Background of the Present Disclosure
FIELD OF DISCLOSURE
[0001] The present disclosure relates to the technical field of biology, and particularly,
to a guide RNA, a CRISPR/AcC2C9 gene editing system, and a gene editing method.
DESCRIPTION OF RELATED ARTS
[0002] Clustered regularly interspaced short palindromic repeats (CRISPR) and the associated
proteins (Cas), initially discovered as adaptive immune systems in archaea and bacteria,
have now developed into multifunctional genome editing tools due to the nuclease activity
of their effector proteins. A guide RNA directs a Cas nuclease to specific targets
in the genome of multiple cells, then the Cas nuclease performs cleavage to cause
DNA double-stranded breakage, and later the endogenous or exogenous DNA repair mechanism
of the cell is carried out, thus achieving the target gene modification. Genomic editing
technology can promote gene engineering, cell engineering, the construction of model
animals, plant gene screening, etc., and has great potential in the treatment of human
diseases.
[0003] At present, CRISPR/Cas9 and CRISPR/Cas12a are the two most widely used genome editing
systems. However, these two systems, both of which are large proteins of more than
1000 amino acids, have a large size. It is known that such a large molecular weight
is difficult to deliver, thus limiting their use in the field of disease treatment.
Recently, a Cas protein with a small size, namely C2C9, which consists of about 400-700
amino acid residues and contains a RuvC nuclease domain, has been discovered. Such
small-sized proteins can help solve the problem of load limitation of adeno-associated
virus vectors. It was reported that
Actinomadura craniella C2C9 (AcC2C9, 506 amino acids) can target the target gene in eukaryotic cells and
realize gene insertion and deletion with an editing efficiency of only 0.1%-25%. Therefore,
it is particularly urgent to optimize the AcC2C9 gene editing system and make it an
efficient and accurate gene editing tool.
SUMMARY OF THE PRESENT DISCLOSURE
[0004] The present disclosure provides a guide RNA, a CRISPR/AcC2C9 gene editing system
and a gene editing method. The nuclease with a small size in the present disclosure
has a high editing efficiency. By utilizing the optimized gene editing system and
method provided by the present disclosure, efficient and accurate editing of target
genes or target genomes can be realized
in-vitro or
in-vivo (including within cells), and the gene editing efficiency of the small-sized CRISPR/AcC2C9
system is greatly improved.
[0005] The present disclosure provides a guide RNA, comprising an RNA framework and a gene
targeting section, the RNA framework includes a tracrRNA and a tracr duplex linked
directly or through a linker; a nucleotide sequence of the tracrRNA is a sequence
obtained by addition, deletion, or substitution of one or more nucleotides in a nucleotide
sequence set forth in SEQ ID NO. 117, and a nucleotide sequence of the tracr duplex
is a sequence obtained by addition, deletion, or substitution of one or more nucleotides
in a nucleotide sequence set forth in SEQ ID NO. 127.
[0006] The present disclosure also provides an isolated polynucleotide, encoding the guide
RNA as described above.
[0007] The present disclosure also provides a construct, containing the isolated polynucleotide
as described above.
[0008] The present disclosure also provides an expression system, containing the construct
as described above or a genome exogenously introduced with the polynucleotide as described
above.
[0009] The present disclosure also provides a gene editing system, including the guide RNA
or the polynucleotide as described above.
[0010] The present disclosure also provides a composition, including the gene editing system,
and a pharmaceutically acceptable carrier.
[0011] The present disclosure also provides a gene editing method, including contacting
a target gene with the gene editing system as described above to realize edition of
the target gene.
[0012] The present disclosure also provides a use of the guide RNA, the isolated polynucleotide,
the construct, the expression system, the gene editing system, the pharmaceutical
composition or the method as described above in gene editing of a target gene and/or
a related polypeptide thereof in an
in vivo, ex vivo cell or cell-free environment.
[0013] The present disclosure also provides a cell, including those that have been genetically
modified with the AcC2C9 nuclease or a polynucleotide encoding the same, the guide
RNA or a polynucleotide encoding the same, the recombinant expression vector, the
system, or the composition as described above.
[0014] As described above, the guide RNA, the CRISPR/AcC2C9 gene editing system and the
gene editing method of the present disclosure have the following beneficial effects:
- (1) In the present disclosure, the guide RNA of AcC2C9 is thoroughly investigated,
and the guide RNA with the highest editing efficiency is developed by performing several
rounds of guide RNA modification, thus the CRISPR/AcC2C9 system which is an efficient
and accurate genome editing tool is obtained. In the present disclosure, the editing
efficiency of 35 target sequences across 6 endogenous genes is tested in a mammalian
cell genome editing experiment, and the results show that the optimal version gRNA_M9
exhibits an enhanced gene editing efficiency across all the tested target sites. Compared
with the original version gRNA_M1, the editing efficiency of 32 target sites of the
optimized gRNA_M9 is about 2-10 times higher.
- (2) The optimized version gRNA_M9 of the present disclosure enables the CRISPR/AcC2C9
system to exhibit editing activity at certain target sites where no activity is detected
when using the original version gRNA_M1. Among the 35 tested target sites, there are
3 sites where no gene editing can be detected when using the original version gRNA_M1
as the guide RNA. However, when using the optimal version gRNA_M9, approximately 10%
gene editing efficiency can be observed. This demonstrates that the guide RNA of the
present disclosure improves the applicability of the small-sized CRISPR system in
cellular gene editing.
- (3) The guide RNA in the present disclosure has a significantly smaller size than
conventional guide RNA, and after engineering the AcC2C9 system also has a small size,
thus providing more possibilities for gene editing and gene therapy which are based
on AAV transmission. This compact and small-sized CRISPR system has a broad application
prospect in genome editing.
BRIEF DESCRIPTION OF THE DRAWINGS
[0015]
Fig. 1 is a schematic diagram showing the specific modification sites and corresponding
base lengths for engineering of the original version guide RNA (gRNA_M1) and the optimized
version guide RNA (M2-M14) in the present disclosure for the AcC2C9 nuclease in Example
1.
Fig. 2 is a diagram of the results of gene editing in mammalian cells mediated by
the AcC2C9 nuclease with different versions of the guide RNA in Example 1. The results
show that the editing efficiency of the partially engineered guide RNA is higher than
that of the original version gRNA_M1, wherein gRNA_M9 has the highest efficiency.
Fig. 3 is a diagram of the results of gene editing in mammalian cells mediated by
the AcC2C9 nuclease with the original version gRNA_M1, as well as the AcC2C9 nuclease
with the optimal version gRNA_M9 in Example 2. The results show that the optimal version
gRNA_M9 greatly improves the gene editing efficiency of a nuclease in mammalian cells;
and the optimal version gRNA_M9 expands the gene editing range of a nuclease in mammalian
cells.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
[0016] The present disclosure provides a guide RNA (gRNA), comprising an RNA framework and
a gene targeting section, wherein the nucleotide sequence of the RNA framework is
shown as SEQ ID NOs. 106-107, or is a sequence obtained by addition, deletion, or
substitution of one or more nucleotides in the nucleotide sequence set forth in SEQ
ID NO. 106 or SEQ ID NO. 107.
[0017] The RNA framework includes a trans-activating CRISPR RNA (tracrRNA) and a tracr duplex.
The tracrRNA and the tracr duplex can be linked directly or through a linker. The
linker is composed of oligonucleotides; preferably, the number of oligonucleotides
in the linker is 3-18 nt; further preferably, in one embodiment, the nucleotide sequence
of the linker is GAAA.
[0018] In terms of the guide RNA described in the present disclosure, the tracr duplex and
the tracrRNA sequence can be linked together to form a single RNA framework, i.e.,
the guide RNA is a single strand, which sequentially comprises the tracr RNA sequence
and the tracr duplex sequence from the 5' end to the 3' end. When the guide RNA is
one strand, the 3' end of the tracr RNA sequence and the 5' end of the tracr duplex
sequence are linked directly or through a linker. When there is no linker, the tracrRNA
sequence + the tracr duplex sequence forms the RNA framework; when there is a linker,
the tracrRNA sequence + the linker + the tracr duplex sequence forms the RNA framework.
[0019] The guide RNA described in the present disclosure comprises sequences obtained by
addition, deletion, or substitution of one or more nucleotides at the 5' or 3' end
of the nucleotide sequence; e.g., sequences obtained by addition, deletion, or substitution
of 15-150 nucleotides at the 5' or 3' end of the nucleotide sequence; or sequences
with deletion of one or more nucleotides in the middle of the nucleotide sequence
or sequences with substitution of 15-150 nucleotides in the middle of the nucleotide
sequence, which are ligated using a linker, wherein the linker is composed of oligonucleotides.
The number of the nucleotides being added, deleted or substituted can be 15-30, 30-45,
45-60, 60-75, 75-90, 90-105, 105-120, 120-135 or 135-150.
[0020] In the guide RNA described in the present disclosure, the gene targeting section
is a nucleotide sequence complementary to the target sequence in the target gene.
The gene targeting section is located at the 3' end of the RNA framework; and identifies
the PAM sequence upstream/downstream of the target sequence; preferably, the PAM sequence
is 5'-NAAN, wherein N is A, T, C, or G, and more preferably, the PAM sequence is 5'-NAAG.
The gene targeting section targets the nucleic acid fragment with a length of 12-40
bp (for example, 13-20, 18-25, 22-32, 26-37, 30-38, or 32-40) downstream the PAM sequence,
preferably, the gene targeting section targets the nucleic acid fragment with a length
of 20 bp downstream the PAM sequence. In the present disclosure, the percentage of
complementarity between the gene targeting section of the guide RNA and the target
sequence in the target gene can be at least 50% (e.g., at least 55%, at least 60%,
at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%,
at least 95%, at least 97%, at least 98%, at least 99%, or 100%). In one embodiment,
the nucleotide sequence of the gene targeting section is set forth in SEQ ID NO. 145.
[0021] The guide RNA described in the present disclosure comprises stem-loop structures
that form a protein binding structure interacting with a nuclease such as AcC2C9.
In some embodiments, the protein binding structure of the guide RNA comprises 6 stem-loop
structures, including stem-loop 1, 2, 3, 4, 5 and 6 (as shown in Fig. 1, Stem1, Stem2,
Stem3, Stem4, Stem5 and Stem6 respectively represent stem-loop 1-6). In some other
embodiments, the protein binding structure of the guide RNA comprises 5 stem-loop
structures, including stem-loop 2, 3, 4, 5 and 6 (i.e., the guide RNA with Stem1 in
Fig. 1 being removed).
[0022] In one embodiment, the nucleotide sequence of the RNA framework is set forth in SEQ
ID NO. 106, wherein the nucleotide sequence of the tracrRNA is set forth in SEQ ID
NO. 117, and the nucleotide sequence of the tracr duplex is set forth in SEQ ID NO.
127. Furthermore, the nucleotide sequence of the guide RNA is set forth in SEQ ID
NO. 4, and this guide RNA is referred to as gRNA_M1.
[0023] In one embodiment, the nucleotide sequence of the RNA framework is obtained by deletion
of 30 bases in stem-loop 1 in the nucleotide sequence set forth in SEQ ID NO. 106.
The nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 107, wherein
the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 140, and the nucleotide
sequence of the tracr duplex is set forth in SEQ ID NO. 127. Further, the nucleotide
sequence of the guide RNA is set forth in SEQ ID NO. 5, and this guide RNA is referred
to as gRNA_M2.
[0024] In one embodiment, the nucleotide sequence of the RNA framework is obtained by deletion
of 34 bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 107.
The nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 108, wherein
the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 118, and the nucleotide
sequence of the tracr duplex is set forth in SEQ ID NO. 128. Further, the nucleotide
sequence of the guide RNA is set forth in SEQ ID NO. 6, and this guide RNA is referred
to as gRNA_M3.
[0025] In one embodiment, the nucleotide sequence of the RNA framework is obtained by deletion
of 43 bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 107,
and the nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 109, wherein
the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 119, and the nucleotide
sequence of the tracr duplex is set forth in SEQ ID NO. 129. Further, the nucleotide
sequence of the guide RNA is set forth in SEQ ID NO. 7, and this guide RNA is referred
to as gRNA_M4.
[0026] In one embodiment, the nucleotide sequence of the RNA framework is obtained by deletion
of 51 bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 107.
The nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 110, wherein
the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 120, and the nucleotide
sequence of the tracr duplex is set forth in SEQ ID NO. 130. Further, the nucleotide
sequence of the guide RNA is set forth in SEQ ID NO. 8, and this guide RNA is referred
to as gRNA_M5.
[0027] In one embodiment, the nucleotide sequence of the RNA framework is obtained by deletion
of 57 bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 107.
The nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 111, wherein
the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 121, and the nucleotide
sequence of the tracr duplex is set forth in SEQ ID NO. 131. Further, the nucleotide
sequence of the guide RNA is set forth in SEQ ID NO. 9, and this guide RNA is referred
to as gRNA_M6.
[0028] In one embodiment, the nucleotide sequence of the RNA framework is obtained by deletion
of 64 bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 107.
The nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 112, wherein
the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 122, and the nucleotide
sequence of the tracr duplex is set forth in SEQ ID NO. 132. Further, the nucleotide
sequence of the guide RNA is set forth in SEQ ID NO. 10, and this guide RNA is referred
to as gRNA_M7.
[0029] In one embodiment, the nucleotide sequence of the RNA framework is obtained by deletion
of 70 bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 107.
The nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 113, wherein
the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 123, and the nucleotide
sequence of the tracr duplex is set forth in SEQ ID NO. 133. Further, the nucleotide
sequence of the guide RNA is set forth in SEQ ID NO. 11, and this guide RNA is referred
to as gRNA_M8.
[0030] In one embodiment, the nucleotide sequence of the RNA framework is obtained by deletion
of 74 bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 107.
The nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 114, wherein
the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 124, and the nucleotide
sequence of the tracr duplex is set forth in SEQ ID NO. 134. Further, the nucleotide
sequence of the guide RNA is set forth in SEQ ID NO. 12, and this guide RNA is referred
to as gRNA_M9.
[0031] In one embodiment, the nucleotide sequence of the RNA framework is obtained by deletion
of 78 bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 107.
The nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 115, wherein
the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 125, and the nucleotide
sequence of the tracr duplex is set forth in SEQ ID NO. 135. Further, the nucleotide
sequence of the guide RNA is set forth in SEQ ID NO. 13, and this guide RNA is referred
to as gRNA_M10.
[0032] In one embodiment, the nucleotide sequence of the RNA framework is obtained by deletion
of 82 bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 107.
The nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 116, wherein
the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 126, and the nucleotide
sequence of the tracr duplex is set forth in SEQ ID NO. 136. Further, the nucleotide
sequence of the guide RNA is set forth in SEQ ID NO. 14, and this guide RNA is referred
to as gRNA_M11.
[0033] In some embodiments of the present disclosure, the guide RNA further includes an
RNA structure stabilizing sequence provided at the 3' end of the guide RNA.
[0034] In some embodiments of the present disclosure, the RNA structure stabilizing sequence
is set forth in SEQ ID NOs. 137-139.
[0035] In one embodiment, the RNA structure stabilizing sequence TTTTATTTTTT is provided
adjacent to the gene targeting section of gRNA_M1 to obtain gRNA_M12. Further, the
nucleotide sequence of the gRNA_M12 is set forth in SEQ ID NO. 15.
[0036] In one embodiment, the RNA structure stabilizing sequence TTGACGCGGTTCTATCTAGTTACGCGTTAAACCAACTAGAAA
is provided adjacent to the gene targeting section of gRNA_M1 to obtain gRNA_M13.
The nucleotide sequence of the gRNA_M13 is set forth in SEQ ID NO. 16.
[0037] In one embodiment, the RNA structure stabilizing sequence ACATGCGATTGACGCGGTTCTATCTAGTTACGCGTTAAACCAACTAGAAA
is provided adjacent to the gene targeting section of gRNA_M1 to obtain gRNA_M14.
The nucleotide sequence of the gRNA_M14 is set forth in SEQ ID NO. 17.
[0038] In some embodiments, the guide RNA further comprises a transcription terminator.
[0039] In the guide RNA described in the present disclosure, the gene targeting section
comprises a nucleotide sequence complementary to a target sequence in a target gene.
The target sequence of the target gene hybridizes with (i.e., base pairing) the gene
targeting section in a sequence-specific manner. The gene targeting section of the
guide RNA can be modified by, for example, genetic engineering, so that the guide
RNA hybridizes with any desired sequence within the target gene. The guide RNA guides
the bound polypeptide to a specific nucleotide sequence in the target gene through
the gene targeting section.
[0040] In some embodiments, the target gene is a DNA sequence. In some embodiments, the
target gene is an RNA sequence.
[0041] The present disclosure further provides a method for modification of the guide RNA.
The variant sequence of the guide RNA refers to a sequence obtained by addition, deletion,
or substitution of one or more nucleotides in the nucleotide sequences set forth in
SEQ ID NO. 4-SEQ ID NO. 5. Preferably, the variant sequence of the guide RNA refers
to a sequence obtained by deletion of nucleotides at the 5' end and/or 3' end of the
nucleotide sequences set forth in SEQ ID NO. 4-SEQ ID NO. 5, i.e., a certain number
of nucleotides can be deleted or truncated at the 5' end only, a certain number of
nucleotides can be deleted or truncated at the 3' end only, or a certain number of
nucleotides can be deleted or truncated at the 5' end and the 3' end simultaneously.
Preferably, the variant sequence of the guide RNA refers to a sequence obtained by
truncation, addition or substitution of nucleotides within the nucleotide sequences
set forth in SEQ ID NO. 4-SEQ ID NO. 5 (other than 5' end and/or 3' end), i.e., any
combination of truncation, addition and substitution of nucleotides at any one or
more positions in the middle of the nucleotide sequence. Preferably, the variant sequence
of the guide RNA refers to a sequence obtained by addition, deletion, or substitution
of one or more nucleotides at the 5' end and/or the 3' end and/or any position in
the middle of the nucleotide sequences set forth in SEQ ID NOs. 4-5.
[0042] In one embodiment, the variant sequence of the guide RNA is obtained by deletion
of 30 bases in stem-loop 1 in the nucleotide sequence set forth in SEQ ID NO. 4, and
the nucleotide obtained by the deletion is referred to as gRNA_M2; the nucleotide
sequence of gRNA_M2 is set forth in SEQ ID NO. 5.
[0043] In one embodiment, the variant sequence of the guide RNA is obtained by deletion
of 34 nt bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 5,
and a GAAA linker is added at the truncated position to obtain gRNA_M3; the nucleotide
sequence of gRNA_M3 is set forth in SEQ ID NO. 6.
[0044] In one embodiment, the variant sequence of the guide RNA is obtained by deletion
of 43 nt bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 5,
and a GAAA linker is added at the truncated position to obtain gRNA_M4; the nucleotide
sequence of gRNA_M4 is set forth in SEQ ID NO. 7.
[0045] In one embodiment, the variant sequence of the guide RNA is obtained by deletion
of 51 nt bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 5,
and a GAAA linker is added at the truncated position to obtain gRNA_M5; the nucleotide
sequence of gRNA_M5 is set forth in SEQ ID NO. 8.
[0046] In one embodiment, the variant sequence of the guide RNA is obtained by deletion
of 57 nt bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 5,
and a GAAA linker is added at the truncated position to obtain gRNA_M6; the nucleotide
sequence of gRNA_M6 is set forth in SEQ ID NO. 9.
[0047] In one embodiment, the variant sequence of the guide RNA is obtained by deletion
of 64 nt bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 5,
and a GAAA linker is added at the truncated position to obtain gRNA_M7; the nucleotide
sequence of gRNA_M7 is set forth in SEQ ID NO. 10.
[0048] In one embodiment, the variant sequence of the guide RNA is obtained by deletion
of 70 nt bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 5,
and a GAAA linker is added at the truncated position to obtain gRNA_M8; the nucleotide
sequence of gRNA_M8 is set forth in SEQ ID NO. 11.
[0049] In one embodiment, the variant sequence of the guide RNA is obtained by deletion
of 74 nt bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 5,
and a GAAA linker is added at the truncated position to obtain gRNA_M9; the nucleotide
sequence of gRNA_M9 is set forth in SEQ ID NO. 12.
[0050] In one embodiment, the variant sequence of the guide RNA is obtained by deletion
of 78 nt bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 5,
and a GAAA linker is added at the truncated position to obtain gRNA_M10; the nucleotide
sequence of gRNA_M10 is set forth in SEQ ID NO. 13.
[0051] In one embodiment, the variant sequence of the guide RNA is obtained by deletion
of 82 nt bases in stem-loop 6 in the nucleotide sequence set forth in SEQ ID NO. 5,
and a GAAA linker is added at the truncated position to obtain gRNA_M11; the nucleotide
sequence of gRNA_M11 is set forth in SEQ ID NO. 14.
[0052] In one embodiment, the variant sequence of the guide RNA is obtained by addition
of TTTTATTTTTT at the 3' end of the nucleotide sequence set forth in SEQ ID NO. 4
and this variant sequence of the guide RNA is referred to asgRNA_M12; the nucleotide
sequence of gRNA_M12 is set forth in SEQ ID NO. 15.
[0053] In one embodiment, the variant sequence of the guide RNA is obtained by addition
of TTGACGCGGTTCTATCTAGTTACGCGTTAAACCAACTAGAAA at the 3' end of the nucleotide sequence
set forth in SEQ ID NO. 4 and this variant sequence of the guide RNA is referred to
as gRNA_M13; the nucleotide sequence of gRNA_M13 is set forth in SEQ ID NO. 16.
[0054] In one embodiment, the variant sequence of the guide RNA is obtained by addition
of
ACATGCGATTGACGCGGTTCTATCTAGTTACGCGTTAAACCAACTAGAAA at the 3' end of the nucleotide
sequence set forth in SEQ ID NO. 4 and this variant sequence of the guide RNA is referred
to as gRNA_M14; the nucleotide sequence of gRNA_M14 is set forth in SEQ ID NO. 17.
[0055] The present disclosure also provides a modified guide RNA, which can achieve hybridization
with any desired sequence in the target gene through modification; or, the properties
of which can be altered through modification, such as enhancing the stability of the
guide RNA through modification, including but not limited to prolonging its half-life
in cells by increasing its resistance to degradation caused by ribonucleases (RNases)
in cells; or, which can enhance the formation or stability of the CRISPR-AcC2C9 genome
editing complex comprising the guide RNA and an endonuclease (e.g., the AcC2C9 nuclease)
through modification; or, which can enhance the specificity of the genome editing
complex through modification; or, which can enhance the initiation site, stability,
or kinetics of the interaction between the genome editing complex and the target sequence
in the genome through modification; or, which can reduce the likelihood or extent
of innate immune responses triggered by the RNA introduced into the cell through modification.
In the present disclosure, multiple properties of the CRISPR-AcC2C9 system (as described
below) can be altered through modifications to the guide RNA, such as enhancing the
formation, on-target activity, specificity, stability, or kinetic characteristics
of the CRISPR-AcC2C9 genome editing complex. RNA can be modified using known methods
in the art, including but not limited to 2'-fluoro, 2'-amino modifications, etc. on
the ribose of pyrimidine, base residues, or the inverted bases at the 3' end of RNA.
In the present disclosure, the guide RNA can undergo any one modification or a combination
of multiple modifications. In some embodiments, the guide RNA introduced into the
cell is modified to edit the loci of any one or more genomes.
[0056] The present disclosure also provides an isolated polynucleotide, encoding any of
the guide RNA as described above.
[0057] The present disclosure also provides a construct, containing the isolated polynucleotide
as described above. The construct can typically be obtained by inserting the isolated
polynucleotide into a suitable expression vector for construction, and a skilled person
in the art can select a suitable expression vector. The construct, for example, can
be a recombinant expression vector, and any suitable expression vector can be used
as long as it is compatible with the host cell, including but not limited to viral
vectors (e.g., poxvirus-based viral vectors; poliovirus; adenovirus; adeno-associated
virus; SV40; herpes simplex virus; human immunodeficiency virus); retroviral vectors
(e.g., murine leukemia virus, spleen necrosis virus, and vectors derived from retroviruses,
e.g., Rous sarcoma virus, Harvey sarcoma virus, avian leukosis virus, lentivirus,
human immunodeficiency virus, myeloproliferative sarcoma virus, and mammary tumor
virus), etc.
[0058] In certain embodiments, multiple guide RNAs are used simultaneously in the same cell
to regulate transcription at different positions on the same target gene or different
target genes. When multiple guide RNAs are used simultaneously, they can exist on
the same expression vector or different vectors, and can also be expressed simultaneously;
when they exist on the same vector, they can be expressed under the same control element.
[0059] In certain embodiments, the nucleotide sequence encoding the guide RNA is operably
linked to a control element, e.g., a transcription control element, for example, a
promoter. In certain embodiments, the nucleotide sequence encoding the guide RNA is
operably linked to an inducible promoter. In certain embodiments, the nucleotide sequence
encoding the guide RNA is operably linked to a constitutive promoter. A transcription
control element can function in eukaryotic cells, e.g., mammalian cells (HEK293T cells);
or prokaryotic cells (e.g., bacterial or archaeal cells). In certain embodiments,
the nucleotide sequence encoding the guide RNA is operably linked to multiple control
elements, allowing the expression of the nucleotide sequence encoding the guide RNA
in both prokaryotic and eukaryotic cells.
[0060] In the present disclosure, the guide RNA can be synthesized by artificial synthesis
methods, for example, by chemical methods, so that it can be easily modified in multiple
ways. Any modification method known in the art can be used, for example, polyA tailing,
addition of a 5' cap analogue, inclusion of a 5' or 3' untranslated region (UTR),
modification of the 5' or 3' end with thiophosphorylated 2'-O-methyl base, or phosphatase
treatment to remove 5' end phosphate ester, etc.
[0061] In some embodiments, the nucleotide sequence encoding the guide RNA comprises one
or more modifications, which can, for example, enhance activity, stability or specificity,
alter delivery, reduce the innate immune response in host cells or for other enhancements.
[0062] In some embodiments, one or more targeting moieties or conjugates that enhance the
activity, cell distribution or cell uptake of the nucleotide sequence encoding the
guide RNA are chemically linked to the guide RNA. The targeting moiety or conjugate
may include a conjugate group covalently bound to a functional group; the conjugate
group includes reporter molecules, polyamines and polyethylene glycol. In some embodiments,
groups that enhance pharmacodynamic properties are linked to gRNA, and the groups
include those improve uptake, enhance resistance to degradation, and/or enhance sequence-specific
hybridization with a target nucleic acid.
[0063] In the present disclosure, the nucleic acid comprising the polynucleotide encoding
the guide RNA may be a nucleic acid mimics. For example, polynucleotide mimics peptide
nucleic acids with good hybridization properties, etc.
[0064] In the present disclosure, the guide RNA or the polynucleotide encoding the guide
RNA is suitable for any biological or in-vitro environment, including but not limited
to bacteria, archaea, fungi, protozoa, plants and animals. Accordingly, suitable target
cells include, but are not limited to, bacterial cells, archaea cells, fungal cells,
protozoan cells, plant cells and animal cells. Suitable target cells can be any type
of cell, including stem cells, somatic cells, etc.
[0065] The present disclosure also provides an expression system, containing the construct
as described above or a genome exogenously introducing the polynucleotide as described
above. The host cell of the expression system is selected from eukaryotic cells and
prokaryotic cells; preferably, the host cell is selected from mouse cells and human
cells.
[0066] The present disclosure also provides a gene editing system, including the guide RNA
or the polynucleotide as described above; further, it may also include a nuclease
or a polynucleotide encoding the same.
[0067] In the editing system described in the present disclosure, the polynucleotide encoding
the nuclease includes: an encoding sequence encoding the nuclease only; an encoding
sequence of the nuclease and various additional encoding sequences; an encoding sequence
of the nuclease (and any additional encoding sequences) and a non-encoding sequence.
The polynucleotide encoding the guide RNA includes: an encoding sequence encoding
the guide RNA only; an encoding sequence of the guide RNA and various additional encoding
sequences; an encoding sequence of the guide RNA (and any additional encoding sequences)
and a non-encoding sequence. In some embodiments, the gene editing system comprises
one or more vectors; the one or more vectors comprise (i) a first regulatory element
operably linked to the polynucleotide encoding the nuclease; and (ii) a second regulatory
element operably linked to the polynucleotide encoding the guide RNA nucleotide sequence;
the (i) and (ii) are on the same vector or different vectors. In some embodiments,
the gene editing system comprises (i) a nuclease or a variant thereof, and (ii) a
vector comprising the sequence encoding the guide RNA. In another embodiment, the
system comprises a complex of the guide RNA and the nuclease.
[0068] The first regulatory element can regulate the transcription of the polynucleotide
encoding the nuclease or its variant. The polynucleotide encoding the nuclease or
its variants may be one or more, and the first regulatory element may be one or more.
The second regulatory element can regulate the transcription of the polynucleotide
encoding the guide RNA. The polynucleotide encoding the guide RNA may be one or more,
and the second regulatory element may be one or more.
[0069] The system described in the present disclosure can comprise one guide RNA or comprise
multiple guide RNAs simultaneously. In one embodiment, the system comprises multiple
guide RNAs to modify different positions on the same target DNA or different target
DNAs simultaneously. In one embodiment, two or more guide RNAs target the same gene
or transcript or loci. In one embodiment, two or more guide RNAs target different
unrelated locus. In some embodiments, two or more guide RNAs target different but
related locus.
[0070] In the gene editing system described in the present disclosure, the nuclease is a
CRISPR nuclease; preferably, the nuclease is selected from Cas9, Cas12, Cas13 protein
families and variants thereof; further preferably, the Cas nuclease is selected from
nSpCas9 and a variant thereof, SaCas9 and a variant thereof, Cas12a and a variant
thereof, as well as C2C9 and a variant thereof; more preferably, the Cas nuclease
is an AcC2C9 nuclease or a variant thereof. In some embodiments, the AcC2C9 nuclease
is directly provided as a protein; for example, acting as an exogenous protein to
transform spheroplast and/or using nucleic acid of the AcC2C9 nuclease to transform
fungi. The AcC2C9 nuclease can be introduced into a cell by any suitable method, such
as injection and the like. The gene editing system described in the present disclosure
identifies the PAM sequence upstream of the target sequence; preferably, the PAM sequence
is 5'-NAAN, wherein N is A, T, C, or G; and more preferably, the PAM sequence is 5'-NAAG.
The gene editing system targets a nucleic acid fragment with a length of 12-40 bp
(such as 20 bp) downstream of the PAM sequence. The gene editing system targets at
least one target sequence in the cell genome.
[0071] In certain embodiments, the nucleic acid encoding the AcC2C9 nuclease is DNA. In
certain embodiments, the nucleic acid encoding the AcC2C9 nuclease is RNA. In certain
embodiments, the nucleic acid encoding the AcC2C9 nuclease is an expression vector,
e.g., a recombinant expression vector. Any suitable expression vector can be used
as long as it is compatible with the host cell, including but not limited to viral
vectors (e.g., poxvirus-based viral vectors; poliovirus; adenovirus; adeno-associated
virus; SV40; herpes simplex virus; human immunodeficiency virus); retroviral vectors
(e.g., murine leukemia virus, spleen necrosis virus, and vectors derived from retroviruses,
e.g., Rous sarcoma virus, Harvey sarcoma virus, avian leukosis virus, lentivirus,
human immunodeficiency virus, myeloproliferative sarcoma virus, and mammary tumor
virus), etc.
[0072] In one embodiment, the nucleic acid encoding the AcC2C9 nuclease is set forth in
SEQ ID NO. 2. In one embodiment, the present disclosure provides a codon-optimized
polynucleotide sequence of the AcC2C9 nuclease, which has at least 90%, 92%, 93%,
94%, 95%, 96%, 97%, 98%, 99%, 99.2%, 99.5%, 99.8%, 99.9%, or 100% sequence homology
to SEQ ID NO: 2. In a preferred embodiment, the human codon-optimized sequence encoding
the AcC2C9 nuclease is set forth in SEQ ID NO. 3, which encodes one or more functional
AcC2C9 domains or a polypeptide having the same function as the polypeptide encoded
by the original natural nucleotide sequence.
[0073] In certain embodiments, the nucleotide sequence encoding the AcC2C9 nuclease is operably
linked to a control element, e.g., a transcription control element, for example, a
promoter. In certain embodiments, the nucleotide sequence encoding the AcC2C9 nuclease
is operably linked to an inducible promoter. In certain embodiments, the nucleotide
sequence encoding the AcC2C9 nuclease is operably linked to a constitutive promoter.
A transcription control element can function in eukaryotic cells, e.g., mammalian
cells (HEK293T cells); or prokaryotic cells (e.g., bacterial or archaeal cells). In
certain embodiments, the nucleotide sequence encoding the AcC2C9 nuclease is operably
linked to multiple control elements, allowing the expression of the nucleotide sequence
encoding the AcC2C9 nuclease in both prokaryotic and eukaryotic cells. In some embodiments,
the polynucleotide sequence encoding the AcC2C9 nuclease is operably linked to a suitable
nuclear localization signal for expression in a cellular or in-vitro environment.
[0074] In the present disclosure, the polynucleotide encoding the AcC2C9 nuclease can be
synthesized by artificial synthesis methods, for example, by chemical methods, so
that it can be easily modified in multiple ways. Any modification method known in
the art can be used. In some embodiments, the polynucleotide encoding the AcC2C9 nuclease
comprises one or more modifications, e.g., modifications for enhancing transcription
activity, changing enzyme activity, improving its translation or stability (e.g.,
increasing its resistance to proteolysis and degradation) or specificity, changing
solubility, changing delivery, and reducing innate immune response in host cells.
The modification can use any modification method known in the art. In some embodiments,
the DNA or RNA encoding the AcC2C9 nuclease introduced into the cell is modified to
edit the loci of any one or more genomes. In some embodiments, the nucleic acid sequence
encoding the AcC2C9 nuclease is a modified nucleic acid, e.g., a codon-optimized nucleic
acid. The modification may be a single modification or a combination modification.
[0075] In the present disclosure, the nucleic acid comprising the polynucleotide encoding
the AcC2C9 nuclease may be a nucleic acid mimics. For example, polynucleotide mimics
peptide nucleic acids with good hybridization properties, etc.
[0076] In the present disclosure, the AcC2C9 nuclease or the polynucleotide encoding the
AcC2C9 nuclease is suitable for any biological or in-vitro environment, including
but not limited to bacteria, archaea, fungi, protozoa, plants or animals. Accordingly,
suitable target cells include, but are not limited to, eukaryotic cells and prokaryotic
cells, e.g., bacterial cells, archaea cells, fungal cells, protozoan cells, plant
cells or animal cells; the eukaryotic cells comprise mammalian cells and plant cells,
and the prokaryotic cells include
E.
coli and
Klebsiella pneumoniae. Suitable target cells can be any type of cell, including stem cells, somatic cells,
etc. The present disclosure is preferably used for the mammalian cell HEK293T cell.
The cells can be
in vivo or
ex vivo. In some embodiments, the AcC2C9 nuclease or the nucleic acid encoding the AcC2C9
nuclease is formulated in liposomes or lipid nanoparticles.
[0077] In the system described in the present disclosure, the AcC2C9 nuclease and the guide
RNA can form a complex in a host cell, and recognize a PAM sequence upstream of a
target gene (such as a target DNA) sequence; the target sequence of the CRISPR/AcC2C9
gene editing system is a nucleic acid fragment (such as a DNA fragment) with a length
of 20 bp downstream the PAM sequence. In one embodiment, the complex can selectively
regulate the transcription of a target DNA in a host cell. The CRISPR/AcC2C9 gene
editing system can cleave the double strand of the target DNA, resulting in DNA breakage.
[0078] In one embodiment, the system comprises a recombinant expression vector. In one embodiment,
the system comprises a recombinant expression vector comprising (i) a nucleotide sequence
encoding a guide RNA, wherein the guide RNA comprises: (a) a first section comprising
a nucleotide sequence complementary to a sequence in a target DNA; and (b) a second
section interacting with the AcC2C9 nuclease; and (ii) a nucleotide sequence encoding
the AcC2C9 nuclease, wherein the AcC2C9 nuclease comprises: (a) an RNA-binding domain
which interacts with the guide RNA; and (b) a domain which regulates transcription
of the target DNA, wherein the site for regulating transcription in the target DNA
is determined by the guide RNA.
[0079] In the present disclosure, an AcC2C9 nuclease variant can also be formed through
modification, mutation, DNA shuffling, etc., so that the AcC2C9 nuclease variant has
improved and desired characteristics, e.g., function, activity, kinetics, half-life,
etc. The modification can be, for example, deletion, insertion or substitution of
amino acids, and can also be, for example, replacement of the "cleavage domain" of
AcC2C9 nucleases with homologous or heterologous cleavage domains from other different
nucleases (e.g., the HNH domain of a CRISPR-related nuclease); proteins can be modified
by any method based on DNA binding and/or DNA modifying known in the art, such as
methylation, demethylation, acetylation, etc., so that for example, the DNA targeting
of the AcC2C9 nuclease can be altered. The DNA shuffling refers to exchanging sequence
fragments between DNA sequences of AcC2C9 nucleases from different sources, to generate
chimeric DNA sequences encoding synthetic proteins with RNA-directed endonuclease
activity. The modification, mutation, DNA shuffling, etc. can be used singly or in
combination.
[0080] Specifically, the AcC2C9 nuclease described in the present disclosure can be:
- (I) a wild-type AcC2C9 nuclease or a fragment thereof which is directed by RNA and
has a nucleic acid binding activity towards RNA; the AcC2C9 nuclease is derived from
Actinomadura craniellae C2C9, with an amino acid sequence set forth in SEQ ID NO. 1; preferably, a humanized
codon-optimized nucleic acid sequence of the AcC2C9 nuclease is set forth in SEQ ID
NO. 3;
- (II) a variant having at least 50% sequence homology with the amino acid sequence
in (I) and having a nucleic acid binding activity towards RNA and being directed by
RNA;
- (III) based on (I) or (II), the AcC2C9 nuclease further includes a nuclear localization
signal fragment;
- (IV) based on (I) or (II) or (III), the AcC2C9 nuclease further comprises:
- (a) one or more modifications or mutations, which result in a significantly reduced
endonuclease activity or a loss of endonuclease activity; and/or
- (b) polypeptides or domains with other functional activities; and
- (V) based on (I) or (II) or (III), the AcC2C9 nuclease has an endonuclease activity.
[0081] In some embodiments, the AcC2C9 can be used in combination with other enzymes or
other components to further develop various potential applications of the AcC2C9 nuclease.
Non-limiting examples of the AcC2C9 nuclease variant in (IV) include for example,
a single-gene editing system based on the AcC2C9 nuclease by fusing inactivated AcC2C9
with a base deaminase; a Prime editing system based on the AcC2C9 nuclease by fusing
inactivated AcC2C9 with a reverse transcriptase; a transcription activation system
based on the AcC2C9 nuclease by fusing inactivated AcC2C9 with a transcription activation
factor; an epigenetic modification system based on the AcC2C9 nuclease by fusing inactivated
AcC2C9 with a nucleic acid epigenetic modifying enzyme; and a transcription repression
system based on the AcC2C9 nuclease by utilizing inactivated AcC2C9.
[0082] Specific properties of the AcC2C9 nuclease variant are as follows, including but
not limited to:
having enhanced or reduced ability to bind to target sites, or retaining ability to
bind to target sites;
having enhanced or reduced endoribonuclease and/or endonuclease activity, or retaining
endoribonuclease and/or endonuclease activity;
having deaminase activity, which can act on cytosine, guanine, or adenine bases, followed
by the deaminated site replication and repair within a cell, generating guanine, thymine,
and guanine, respectively;
having ability to regulate transcription of a target DNA, either increasing or decreasing
transcription of the target DNA at specific positions;
altering DNA targeting;
having increased, decreased, or maintained stability;
having ability to cleave the complementary strand of a target DNA, but having reduced
ability to cleave the non-complementary strand;
having ability to cleave the non-complementary strand of a target DNA, but having
reduced ability to cleave the complementary strand;
having reduced ability to cleave both the complementary and non-complementary strands
of a target DNA;
having enzymatic activity to modify polypeptides (e.g., histone) associated with DNA,
wherein the enzymatic activity can be one or more of methyltransferase activity, demethylase
activity, acetyltransferase activity, deacetylase activity, kinase activity, phosphatase
activity, ubiquitin ligase activity, deubiquitinating activity, ribosylation activity,
etc. (covalent modification of proteins is catalyzed by these enzyme activities; for
example, the AcC2C9 nuclease variants modify histone through methylation, acetylation,
ubiquitination, phosphorylation, etc., to induce structural changes in histone-associated
DNA, thereby controlling the structure and properties of DNA).
[0083] In some embodiments, the AcC2C9 nuclease variant has no cleavage activity. In some
embodiments, the AcC2C9 nuclease variant has single-strand cleavage activity. In some
embodiments, the AcC2C9 nuclease variant has double-strand cleavage activity.
[0084] Having enhanced activity or ability refers to the activity or ability being improved
by at least 1%, 5%, 10%, 20%, 30%, 40% or 50% relative to the wild-type AcC2C9 nuclease.
[0085] Having reduced activity and ability refers to the activity or ability is less than
50%, 40% lower than, 30% lower than, 20% lower than, 10% lower than, 5% lower than
or 1% lower than that of the wild-type AcC2C9 nuclease.
[0086] These small-sized AcC2C9 and variants thereof described in the present disclosure
can be used in any system, composition, kit and method described in the present disclosure
below.
[0087] Unless otherwise specified, the terms "AcC2C9" and " AcC2C9 nuclease" include the
wild-type AcC2C9 nuclease and all variants thereof, and those skilled in the art can
determine the types of AcC2C9 nuclease variants by conventional methods which are
not limited to those listed above.
[0088] Each component in the system described in the present disclosure can be transported
by carriers. For example, for polynucleotides, methods that can be used include, but
are not limited to, nanoparticles, liposomes, ribonucleoprotein, small molecule RNA-conjugates,
chimeras, RNA-fusion protein complexes, etc.
[0089] The system described in the present disclosure may further include one or more donor
templates. In certain embodiments, the donor template comprises a donor sequence for
inserting into a target gene.
[0090] The system described in the present disclosure may further comprise a dimeric FOK1
nuclease, and an intact or partially or completely defective AcC2C9 nuclease or a
guide RNA is linked to the dimeric FOK1 nuclease to direct endonuclease cleavage after
being guided to one or more specific DNA target sites using one or more guide RNA
molecules.
[0091] The system described in the present disclosure can edit or modify DNA at multiple
positions in cells for gene therapy, including but not limited to gene therapy for
diseases, biological research, crop resistance improvement or yield improvement, etc.
[0092] The present disclosure also provides a composition, that comprises one or more of
the AcC2C9 nuclease or a polynucleotide encoding the same, the guide RNA or a polynucleotide
encoding the same, the recombinant expression vector, and the system as described
above, and acceptable carriers, vehicles, etc. The acceptable carriers and vehicles
are for example sterile water or normal saline, stabilizers, excipients, antioxidants
(ascorbic acid, etc.), buffers (phosphoric acid, citric acid, other organic acids,
etc.), preservatives, surfactants (PEG, Tween, etc.), chelating agents (EDTA, etc.),
adhesives, etc. In addition, the composition can also contain other low molecular
weight polypeptides; proteins such as serum albumin, gelatin or immunoglobulins; amino
acids such as glycine, glutamine, asparagine, arginine and lysine; saccharides (such
as polysaccharides and monosaccharides) or carbohydrates; sugar alcohols such as mannitol
or sorbitol. When preparing aqueous solution for injection, e.g., normal saline, an
isotonic solution containing glucose or other auxiliary drugs, such as D-sorbitol,
D-mannose, D-mannitol and sodium chloride, appropriate solubilizers for example, alcohols
(ethanol, etc.), polyols (propylene glycol, PEG, etc.) and nonionic surfactants (Tween
80 or HCO-50) and the like can be used. In some embodiments, the composition comprises
a guide RNA and a buffer for stabilizing nucleic acids.
[0093] The present disclosure also provides a kit, including the system or composition as
described above. The kit may further include one or more of, for example, a dilution
buffer; a washing buffer; a control reagent, etc. In some embodiments, the kit includes
(a) the AcC2C9 nuclease as described above or a nucleic acid encoding the AcC2C9 nuclease;
and (b) a guide RNA or a nucleic acid encoding the guide RNA, wherein the guide RNA
can direct the AcC2C9 nuclease or a variant thereof to a target polynucleotide sequence.
In certain embodiments, the kit further contains a donor template comprising a heterologous
polynucleotide sequence, wherein the heterologous polynucleotide sequence can be inserted
into the target polynucleotide sequence.
[0094] The present disclosure also provides a gene editing method, including contacting
a target gene with the gene editing system as described above to realize editing of
the target gene. The method of the present disclosure can be used to target, edit,
modify or manipulate a target gene (such as a target DNA) in cells, in
in vivo, in
ex vivo cell, or in cell-free system, and the method includes: introducing the AcC2C9 nuclease
or a polynucleotide encoding the same, the guide RNA or a polynucleotide encoding
the same, the recombinant expression vector, the system, the composition, etc., into
a kit to target, edit, modify or manipulate the target gene in
in vivo, ex vivo cell or cell-free system. In one embodiment, the method includes the following steps:
- (a) introducing the AcC2C9 nuclease or the nucleic acid encoding the AcC2C9 nuclease
in an in vivo, ex vivo cell or cell-free system;
- (b) introducing the guide RNA (gRNA) or the nucleic acid (e.g., DNA) suitable for
in situ generation of such guide RNA; and
- (c) contacting cells or target genes with the AcC2C9 nuclease or the nucleic acid
encoding the AcC2C9 nuclease, and the guide RNA or the nucleic acid suitable for in
situ generation of such guide RNA, to produce one or more cleavages, nicks or perform
editing in the target gene; wherein the AcC2C9 nuclease is directed to the target
gene by its corresponding processed or unprocessed guide RNA.
[0095] In some embodiments, the gene editing method described in the present disclosure
includes the following steps:
- i) introducing the AcC2C9 nuclease or the polynucleotide encoding the same and the
guide RNA or the polynucleotide encoding the same into a cell;
- ii) under the mediation of the AcC2C9 nuclease, producing one or more nicks in the
target gene, or targeting, editing, modifying or manipulating the target gene.
[0096] Each parameter in the gene editing method described in the present disclosure can
be adjusted according to conventional knowledge in the art. For example, the concentration
of the expression vector including the nuclease and guide RNA is preferably 1 µg;
the editing is preferably 72 hours later the cell transfection.
[0097] In the gene editing method described in the present disclosure, the AcC2C9 nuclease
is guided to a target gene by a processed or unprocessed guide RNA. The AcC2C9 nuclease
and the guide RNA form a complex to recognize a PAM sequence upstream of the target
gene. In some preferred embodiments, the method further includes introducing a donor
template comprising a heterologous polynucleotide sequence into the cell.
[0098] The present disclosure also provides a use of the guide RNA, the isolated polynucleotide,
the construct, the expression system, the gene editing system, the pharmaceutical
composition or the method as described above in gene editing of a target gene and/or
a related polypeptide thereof in an
in vivo, ex vivo cell or cell-free environment. The ex vivo cell is at least one of bacterial cells,
archaea cells, fungal cells, protozoan cells, virus cells, plant cells and animal
cells. The gene editing is selected from the group consisting of gene cleavage, gene
deletion, gene insertion, point mutation, transcription repression, transcription
activation, base editing, and guided editing, including but not limited to:
cleaving a target gene;
manipulating the expression of a target gene;
genetically modifying a target gene;
genetically modifying a target gene-related polypeptide;
intentional and controlled damage at any desired position of a target gene;
intentional and controlled repair at any desired position of the target gene; and
modifying a target gene by means other than introducing double-stranded breakage (the
AcC2C9 nuclease has an enzymatic activity, which allows it to modify a target gene
by means other than introducing double-stranded breakage; the enzymatic activity can
be owned by AcC2C9 itself, or obtained by, for example, fusing a heterologous polypeptide
with an enzymatic activity to the AcC2C9 nuclease to form a chimeric AcC2C9 nuclease,
the enzymatic activity includes but is not limited to methyltransferase activity,
deamination activity, dismutase activity, alkylation activity, demethylase activity,
DNA repair activity, transposase activity, recombinase activity, DNA damage activity,
depurinization activity, oxidation activity, pyrimidine dimer formation activity,
etc.).
[0099] Preferably, the gene editing is gene deletion or gene cleavage; the gene editing
can be used to achieve one or more of the following, including but not limited to
the correction of pathogenic sites, gene function research, cell function enhancement,
cell therapy, etc.
[0100] The AcC2C9 nuclease or a polynucleotide encoding the same, the guide RNA or a polynucleotide
encoding the same, the recombinant expression vector, the system, the composition,
and the kit can be applied to research field, diagnosis field, industry field (e.g.,
microbial engineering), drug development (e.g., high-throughput screening), target
confirmation, imaging field, treatment field, etc.
[0101] In some embodiments, the target gene is a target DNA. In some embodiments, the target
DNA can be a naked DNA
in vitro that is not bound to DNA-related proteins. In some embodiments, the target DNA is
a chromosomal DNA in cells
in vitro. In some embodiments, the target gene is a target RNA. In some embodiments, enabling
the target DNA to contact with a targeting complex comprising the AcC2C9 nuclease
and the guide RNA, and the guide RNA that comprises a nucleotide sequence complementary
to the target DNA provides target specificity for the targeting complex; the AcC2C9
nuclease provides site-specific activity. In some embodiments, the targeting complex
modifies a target DNA, leading to, for example, DNA cleavage, DNA methylation, DNA
damage, DNA repair, etc. In some embodiments, the targeting complex modifies a target
DNA-related polypeptide (e.g., histone, a DNA-binding protein, etc.), thereby leading
to methylation, histone acetylation, histone ubiquitination, etc. of the target DNA-related
polypeptide-histone, for example.
[0102] In the method described in the present disclosure, an AcC2C9 nuclease or a nucleic
acid comprising a nucleotide sequence encoding a polypeptide of the AcC2C9 nuclease
can be introduced into a cell by a commonly known method. Similarly, a guide RNA or
a nucleic acid comprising a nucleotide sequence encoding the guide RNA can be introduced
into a cell by a commonly known method. The commonly known methods include DEAE-dextran
mediated transfection, liposome mediated transfection, virus or phage infection, lipid
transfection, transfection, conjugation, protoplast fusion, polyethyleneimine mediated
transfection, electroporation, calcium phosphate precipitation, gene gun, calcium
chloride precipitation, microinjection, nanoparticle mediated nucleic acid delivery,
etc. For example, electroporation, calcium chloride precipitation, microinjection,
lipid transfection and the like are used for plasmid delivery. For viral vector delivery,
enabling cells to contact with viral particles comprising donor polynucleotides and/or
nucleic acids encoding a guide RNA and/or an AcC2C9 nuclease and/or a chimeric AcC2C9
nuclease.
[0103] In some embodiments, in the method described in the present disclosure, a nuclease
cleaves a target DNA in a cell to generate double-stranded breakage, and then the
cell usually performs repair in the following ways: non-homologous end joining (NHEJ)
and homology-directed repair.
[0104] The present disclosure also provides a cell, wherein the cell includes those have been
genetically modified with the AcC2C9 nuclease or a polynucleotide encoding the same,
the guide RNA or a polynucleotide encoding the same, the recombinant expression vector,
the system and the composition as described above, or those undergo gene editing with
the above gene editing system or the method.
[0105] In the present disclosure, the effective dosage of the guide RNA and/or the AcC2C9
nuclease and/or the recombinant expression vector and/or the donor polynucleotide
is known for those skilled in the art. It can be determined according to different
routes of administration and the characteristics of the disorders to be treated.
[0106] In the present disclosure, the bacteria or prokaryotic bacteria can be
E. coli, Klebsiella pneumoniae, Bacteroides ovatus, Campylobacter jejuni, Staphylococcus
saprophyticus, Enterococcus faecalis, Bacteroides thetaiotaomicron, Bacteroides vulgatus,
Bacteroides uniformis, Lactobacillus casei, Bacteroides fragilis, Acinetobacter Iwoffii,
Fusobacterium nucleatum, Bacteroides johnsonii, Bacteroides arabidopsis, Lactobacillus
rhamnosus, Bacteroides massiliensis, Parabacteroides merdae, Fusobacterium mortiferum, and
Bifidobacterium breve, etc.
[0107] In the present disclosure, the eukaryotic cells include, but are not limited to,
eukaryotic cells such as mammalian cells and fungi. The fungi include yeasts and
Aspergillus, for example,
Saccharomyces cerevisiae, Hansenula polymorpha, Pichia pastoris, Kluyveromyces fragilis,
Kluyveromyces lactis, Schizosaccharomyces pombe, Candida albicans, Candida dubliniensis,
Candida glabrata, Candida guilliermondii, Candida kefyr, Candida krusei, Candida lusitaniae,
Candida melinii, Candida oleophila, Candida parapsilosis, Candida tropicalis, Candida
utilis, Aspergillus fumigatus, Aspergillus flavus, Aspergillus niger, Aspergillus
clavatus, Aspergillus glaucus, Aspergillus nidulans, Aspergillus oryzae, Aspergillus
terreus, Aspergillus ustus, and
Aspergillus versicolor, etc.
[0108] In the examples of the present disclosure, a novel genome editing method based on
a small-sized CRISPR/AcC2C9 nuclease is disclosed. The present disclosure shows that
AcC2C9 can accurately cleave genomic DNA and achieve double-stranded breakage of the
genomic DNA through the guiding function of a guide RNA. The system can efficiently
and accurately achieve gene editing in a living cell by using the host cell's own
or exogenous repair mechanism.
[0109] The present disclosure also provides a method for preparing the guide RNA as described
above, including modification of tracrRNA or crRNA of the RNA framework alone and
modification of tracrRNA and or crRNA of the RNA framework in combination, and the
modification is selected from truncation, addition, or substitution of one or more
nucleotides in tracrRNA or crRNA, or linking tracrRNA and crRNA through a linker to
prepare the guide RNA.
[0110] Parts of the sequences described in the present disclosure are as follows:
The AcC2C9 nuclease described in the present disclosure is Actinomadura craniellae C2C9 (AcC2C9), and its amino acid sequence preferably comprises the sequence shown
below:


E. coli codon-optimized nucleotide sequence encoding the AcC2C9 nuclease comprises the following
sequence:


human codon-optimized nucleotide sequence encoding the AcC2C9 nuclease comprises the
following sequence:


different versions of guide RNA sequences for the AcC2C9 nuclease described in the
present disclosure are shown as follows, in which the light gray shows the RNA framework
sequence, and if there is a linker in the guide RNA, the linker sequence will be GAAA,
with the linker as part of the framework; the dotted underline shows tracrRNA; the double underline shows the sequence of the tracr duplex; the underlined shows sequence of the gene targeting section; the wavy line shows the RNA structure
stabilizing sequence.
[0111] Original version of guide RNA, i.e., gRNA_M1, of CRISPR/AcC2C9 system:
- a schematic diagram of its secondary structure is shown in Fig. 1; wherein the nucleotide
sequence of the RNA framework is set forth in SEQ ID NO. 106, the nucleotide sequence
of the tracrRNA is set forth in SEQ ID NO. 117, and the nucleotide sequence of the
tracr duplex is set forth in SEQ ID NO. 127.
[0112] Optimized version guide RNA sequence gRNA_M2 of CRISPR/AcC2C9 provided in the present
disclosure:

wherein the nucleotide sequence of the RNA framework
is set forth in SEQ ID NO. 107, the nucleotide sequence of the tracrRNA is set forth
in SEQ ID NO. 140, and the nucleotide sequence of the tracr duplex is set forth in
SEQ ID NO. 127.
gRNA_M3:

wherein the
nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 108, the nucleotide
sequence of the tracrRNA is set forth in SEQ ID NO. 118, and the nucleotide sequence
of the tracr duplex is set forth in SEQ ID NO. 128.
|gRNA_M4:

wherein the nucleotide sequence
of the RNA framework is set forth in SEQ ID NO. 109, the nucleotide sequence of the
tracrRNA is set forth in SEQ ID NO. 119, and the nucleotide sequence of the tracr
duplex is set forth in SEQ ID NO. 129.
gRNA_M5:

wherein the nucleotide sequence of the RNA
framework is set forth in SEQ ID NO. 110, the nucleotide sequence of the tracrRNA
is set forth in SEQ ID NO. 120, and the nucleotide sequence of the tracr duplex is
set forth in SEQ ID NO. 130.
gRNA_M6:

wherein the nucleotide sequence of the RNA framework
is set forth in SEQ ID NO. 111, the nucleotide sequence of the tracrRNA is set forth
in SEQ ID NO. 121, and the nucleotide sequence of the tracr duplex is set forth in
SEQ ID NO. 131.
gRNA_M7:

wherein the nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 112,
the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 122, and the nucleotide
sequence of the tracr duplex is set forth in SEQ ID NO. 132.
gRNA_M8:

wherein the nucleotide sequence of the RNA framework is set forth in SEQ
ID NO. 113, the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 123,
and the nucleotide sequence of the tracr duplex is set forth in SEQ ID NO. 133.
gRNA_M9:

wherein the nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 114,
the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 124, and the nucleotide
sequence of the tracr duplex is set forth in SEQ ID NO. 134.
gRNA_M10:

wherein the nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 115,
the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 125, and the nucleotide
sequence of the tracr duplex is set forth in SEQ ID NO. 135.
gRNA_M11:

wherein the
nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 116, the nucleotide
sequence of the tracrRNA is set forth in SEQ ID NO. 126, and the nucleotide sequence
of the tracr duplex is set forth in SEQ ID NO. 136.
gRNA_M12:

wherein the nucleotide sequence of the RNA framework is set forth in SEQ
ID NO. 106, the nucleotide sequence of the tracrRNA is set forth in SEQ ID NO. 117,
the nucleotide sequence of the tracr duplex is set forth in SEQ ID NO. 127, and the
nucleotide sequence of the RNA structure stabilizing sequence is set forth in SEQ
ID NO. 137.
gRNA_M13:

wherein the nucleotide
sequence of the RNA framework is set forth in SEQ ID NO. 106, the nucleotide sequence
of the tracrRNA is set forth in SEQ ID NO. 117, the nucleotide sequence of the tracr
duplex is set forth in SEQ ID NO. 127, and the nucleotide sequence of the RNA structure
stabilizing sequence is set forth in SEQ ID NO. 138.
gRNA_M14:

wherein
the nucleotide sequence of the RNA framework is set forth in SEQ ID NO. 106, the nucleotide
sequence of the tracrRNA is set forth in SEQ ID NO. 117, the nucleotide sequence of
the tracr duplex is set forth in SEQ ID NO. 127, and the nucleotide sequence of the
RNA structure stabilizing sequence is set forth in SEQ ID NO. 139.
[0113] The terms "AcC2C9", "AcC2C9 nuclease", "AcC2C9 polypeptide", and "AcC2C9 protein"
can be used interchangeably.
[0114] The terms "guide RNA", "gRNA", "single gRNA" and "chimeric gRNA" can be used interchangeably.
[0115] The term "a" or "an" entity refers to one or more of such entities; therefore, the
terms "a" (or "an"), "one or more" and "at least one" are used interchangeably herein.
[0116] The term "homology", "identity" or "similarity" refers to the sequence similarity
between two peptides or two nucleic acid molecules. Homology can be determined by
aligning the corresponding positions in different polypeptide or nucleic acid molecules.
When the same position in the compared molecular sequence and another different sequence
is occupied by the same base or amino acid, then the compared molecular sequence and
another different sequence are homologous at that position. The degree of homology
between sequences is determined by the function of the number of matching or positions
shared by the sequences. An "unrelated" or a "non-homologous" sequence should have
less than 20% homology with one of the sequences disclosed in the present disclosure.
[0117] A polynucleotide or polynucleotide region (or polypeptide or polypeptide region)
has a certain percentage (e.g., 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%,
or 99%) of sequence homology with another polynucleotide or polynucleotide region
(or polypeptide or polypeptide region) refers to that the two sequences being aligned
possess that percentage of bases (or amino acids) which are identical. The alignment
and percentage homology or sequence identity can be determined using software procedures
and methods known in the art.
[0118] In the present disclosure, the terms "polynucleotide" and "oligonucleotide" are used
interchangeably, and they refer to polymerized forms of nucleotides (no matter deoxynucleotides
or ribonucleotides or their analogues) of any length. A polynucleotide can have any
three-dimensional structure and can perform any function known or unknown. Examples
of polynucleotides include, but are not limited to, genes or gene fragments (including
probes, primers, EST or SAGE tags), exons, introns, messenger RNA (mRNA), transport
RNA, ribosomal RNA, ribozymes, cDNA, dsRNA, siRNA, miRNA, recombinant polynucleotides,
branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, and isolated
RNA of any sequence, nucleic acid probes, and primers. Polynucleotides also comprise
modified nucleotides, e.g., methylated nucleotides and nucleotide analogues. If there
is a modification of a polynucleotide, the modification can be introduced either before
or after the assembly of the polynucleotide. A nucleotide sequence can be interrupted
by non-nucleotide components. A polynucleotide can be further modified after polymerization,
for example, by coupling to a labeled component. The term refers to both double-stranded
and single-stranded polynucleotide molecules. Unless otherwise specified or required,
any implementation example of a polynucleotide disclosed in the present disclosure
includes its double-stranded form and any one of two complementary single-stranded
forms known or predicted to constitute the double-stranded structure.
[0119] When being applied to a polynucleotide, the term "encode" refers to that the polynucleotide
"encodes" a polypeptide, which means that in its natural state or when being manipulated
by methods commonly known to those skilled in the art, the polynucleotide can be transcribed
and/or translated to produce the polypeptide of interest and/or fragments thereof,
or to produce mRNA that is capable of encoding the polypeptide of interest and/or
fragments thereof. An antisense strand refers to a sequence that is complementary
to the polynucleotide, and from which the encoding sequence can be determined.
[0120] The term "genomic DNA" denotes the DNA of an organism genome, including the DNA of
bacterial, archaeal, fungal, protozoan, viral, plant, or animal genomes.
[0121] The term "manipulating" DNA includes binding, making nicks in one strand, or cleaving
two strands of DNA, or includes modifying or editing DNA or polypeptides that bind
to DNA. Manipulating DNA can silence, activate or regulate the expression of RNA or
the polypeptide encoded by the DNA (prevent transcription, reduce transcription activity,
prevent translation, or lower translation level), or prevent or enhance the binding
of polypeptides and DNA. Cleavage can be carried out by various methods, e.g., enzymatic
or chemical hydrolysis of phosphodiester bonds; it can involve single-strand or double-strand
cleavage; DNA cleavage can result in the generation of blunt ends or staggered ends.
[0122] The term "hybridizable", "complementary" or "substantially complementary" refers
to that a nucleic acid (e.g., RNA) comprises a nucleotide sequence that enables it
to noncovalently bind to another nucleic acid in a sequence-specific and antiparallel
manner under appropriate
in vitro and/or
in vivo temperature and solution ionic strength conditions, i.e., to form Watson-Crick base
pairs and/or G/U base pairs, "anneal," or "hybridize".
[0123] It can be understood in the art that the sequence of a polynucleotide does not need
to be 100% complementary to the sequence of a target nucleic acid to which the polynucleotide
can specifically hybridize. A polynucleotide can hybridize on one or more sections.
A polynucleotide can comprise at least 70%, at least 80%, at least 90%, at least 95%,
at least 99% or 100% sequence complementarity to a target region within the target
nucleic acid sequence.
[0124] The terms "peptide", "polypeptide" and "protein" are used interchangeably in the
present disclosure, and represent polymerized forms of amino acids of any length,
which can include encoded and non-encoded amino acids, chemically or biochemically
modified or derived amino acids, and polypeptides with modified peptide backbones.
[0125] The term the DNA sequence "encoding" a specific RNA is a DNA nucleic acid sequence
transcribed into the RNA. A DNA polynucleotide can encode an RNA that will be translated
into a protein (mRNA), or a DNA polynucleotide can encode an RNA that will not be
translated into a protein (e.g., tRNA, rRNA or gRNA; also known as a "non-coding"
RNA or "ncRNA"). A "protein encoding sequence" or a sequence encoding a specific protein
or polypeptide is a nucleic acid sequence that is transcribed into mRNA (in the case
of DNA) and translated into a polypeptide (in the case of mRNA)
in vivo or
in vitro under the control of an appropriate regulatory sequence.
[0126] The term "vector" or "expression vector" is a replicon, such as a plasmid, phage,
virus or cosmid, to which another DNA section, i.e., an "insertion fragment", can
be attached in order to achieve replication of the section in a cell.
[0127] The term "expression cassette" comprises a DNA encoding sequence operably linked
to a promoter. "Operably linked" denotes parallel linking, with each component in
a relationship that allows them to function in their intended way. The terms "recombinant
expression vector" or "DNA construct" are used interchangeably in the present disclosure
to denote a DNA molecule comprising a vector and at least one insertion fragment.
A recombinant expression vector is usually produced for the purpose of expressing
and/or amplifying inserted fragments or for constructing other recombinant nucleotide
sequences.
[0128] When an exogenous DNA, e.g., a recombinant expression vector, has been introduced
into a cell, the cell has been "genetically modified" or "transformed" or "transfected"
by this DNA. The existence of the exogenous DNA leads to permanent or temporary genetic
changes. The transformed DNA is integrated or not integrated into the genome of the
cell.
[0129] The term "target DNA" is a DNA polynucleotide comprising "target sites" or "target
sequences". The terms "target site", "target sequence", "target protospacer DNA" or
"protospacer-like sequence" are used interchangeably in the present disclosure to
denote the nucleic acid sequence existing in the target DNA, to which the DNA-targeting
section of gRNA will bind if there is suitable condition. RNA molecules comprise sequences
that bind, hybridize or are complementary to a target sequence in a target DNA, thus
directing the linked polypeptide of RNA to a specific position (target sequence) in
the target DNA. "Cleavage" refers to the breakage of the covalent backbone of DNA
molecules.
[0130] The terms "nuclease" and "endonuclease" can be used interchangeably to refer to an
enzyme with endonucleolytic catalytic activity for polynucleotide cleavage. The "cleavage
domain" or "active domain" or "nuclease domain" of a nuclease refers to a polypeptide
sequence or domain with catalytic activity for DNA cleavage within the nuclease. The
cleavage domain can be a single polypeptide chain, or the cleavage activity can be
generated by the association of two or more polypeptides.
[0131] The term "localized polypeptide" or "RNA-binding site-directed polypeptide" refers
to a polypeptide that binds to RNA and targets a specific DNA sequence.
[0132] The term "guide sequence" or DNA-targeting section (or "DNA-targeting sequence")
comprises a nucleotide sequence that is complementary to a specific sequence within
a target DNA (i.e., the complementary strand of the target DNA), with the target DNA
referred to as a "protospacer-like" sequence in the present disclosure.
[0133] The term "recombination" refers to the process of exchanging genetic information
between two polynucleotides. As used in the present disclosure, "homology-directed
repair (HDR)" denotes a specialized form of DNA repair that occurs, for example, during
the repair of double-stranded breakage in cells. This process requires nucleotide
sequence homology, utilizing a "donor" molecule to provide a template for the repair
of a "target" molecule (i.e., the molecule has undergone double-stranded breakage),
and resulting in the transfer of genetic information from the donor to the target.
If the donor polynucleotide is different from the target molecule, and part or all
of the sequences of the donor polynucleotide are incorporated into the target DNA,
the repair directed by homology may lead to changes in the sequence of the target
molecule (e.g., insertion, deletion and mutation).
[0134] The term "non-homologous end joining (NHEJ)" refers to the repair of double-stranded
breakage in DNA by directly joining the broken ends to each other without the need
for a homologous template. NHEJ often leads to the deletion of nucleotide sequences
near the site undergone double-stranded breakage.
[0135] The term "treatment" includes preventing the occurrence of a disease or symptom;
inhibiting a disease or symptom; or alleviating a disease.
[0136] The terms "individual," "subject," "host," and "patient" are used interchangeably
in the present disclosure and denote any mammalian subject, particularly a human,
for whom diagnosis, treatment, or therapy is desired.
[0137] The embodiments of the present disclosure are illustrated below through specific
examples, and those skilled in the art can easily understand other advantages and
effects of the present disclosure from the contents disclosed in this specification.
The present disclosure can also be implemented or applied through other different
detailed embodiments, and various details in this specification can be modified or
changed based on different viewpoints and applications without departing from the
spirit of the present disclosure.
[0138] Before further describing the detailed embodiments of the present disclosure, it
should be understood that the protection scope of the present disclosure is not limited
to the following specific implementation examples; also, it should be understood that
the terms used in the examples of the present disclosure are for the purpose of describing
specific implementation examples, and not for the purpose of limiting the protection
scope of the present disclosure; after reading the present disclosure, those skilled
in the art can make various changes or modifications to the present disclosure, and
these equivalent forms also fall within the scope defined by the claims in the present
disclosure. For example, a single-gene editing system based on AcC2C9 is developed
by fusing an engineered inactivated AcC2C9 with a base deaminase; a Prime editing
system based on AcC2C9 is developed by fusing an inactivated AcC2C9 with a reverse
transcriptase; a transcription activation system based on AcC2C9 is developed by fusing
an inactivated AcC2C9 with a transcription activation factor; an epigenetic modification
system based on AcC2C9 is developed by fusing an inactivated AcC2C9 with a nucleic
acid epigenetic modifying enzyme; and a transcription repression system based on AcC2C9
is developed by utilizing an inactivated AcC2C9. In the specification and claims of
the present disclosure, the singular forms "a", "an" and "the" include the plural
forms unless the context clearly indicates otherwise.
[0139] When the examples give numerical ranges, it should be understood that unless otherwise
specified in the present disclosure, two endpoints of each numerical range and any
numerical value between the two endpoints can be selected. Unless otherwise defined,
all technical and scientific terms used in the present disclosure have the same meaning
as commonly understood by those skilled in the art. In addition to the specific methods,
equipment and materials used in the examples, according to the knowledge of the prior
art by those skilled in the art and the records of the present disclosure, any methods,
equipment and materials in the prior art similar to or equivalent to the methods,
equipment and materials described in the examples of the present disclosure can be
used to achieve the present disclosure.
[0140] In the examples of the present disclosure, the mainly used CRISPR system is the V-U4
type CRISPR system, wherein the effector protein is mainly the
Actinomadura craniellae C2C9 (AcC2C9) nuclease. The AcC2C9 nuclease can be accurately directed to the target
gene under the guidance of the corresponding guide RNA, cleave the genomic DNA, and
achieve the double-stranded breakage of the genomic DNA. The system can efficiently
and accurately achieve gene editing in a living cell by using the host cell's own
or exogenous repair mechanism.
[0141] Where the specific technology or conditions are not specified in the examples, it
should be understood that they are carried out according to the conventional technology
or conditions described in the literature in the art or according to the product manufacturer's
instructions.
[0142] The primers used in the examples were synthesized by Shanghai Sangon Biotech Co.,
Ltd. and Suzhou Genewiz Biotechnology Co., Ltd. Any reagents or instruments used without
specifying the manufacturer are considered products that are commercially available.
Example 1 Engineering of corresponding guide RNAs for AcC2C9 Nuclease
[0143] In this example, the corresponding guide RNAs for the AcC2C9 nuclease were modified
based on an original version gRNA_M1. The specific modification sites and the corresponding
base lengths are illustrated in Fig. 1. The specific engineering methods and the lengths
of the modified bases are shown in the following:
encoding nucleotide of gRNA_M1: 5'-GAACGCGGCCCGGAACATCGAACGCCACGCAGTGCTGATCGATCGAAACGTCG
CCTGCGATAGGCGGGAGACGCTAAACGCCCGTGGAGCATCCATAAGACCAACC ACCTCTCGGGGCGGTAGGCACGACGCATCGAAGCGGGAAGGCTCCGGCGCTC
GGCCTGAGTCACCTCAGCAGAGTGATCTGCTGACGCTCCCAACCTTGAATAACG AAACGGCAACGCCTCCATAGCGGTGCAGGTCAATAAGGGTCGGCCCCACGCGT
GTAGGGAGCGATCGaagtgacagtatcctctgtat-3' (SEQ ID NO. 92)
encoding nucleotide of gRNA_M2, based on M1 version, but with 30 nt bases of stem-loop
1 removed: 5'-CAGTGCTGATCGATCGAAACGTCGCCTGCGATAGGCGGGAGACGCTAAACGCC CGTGGAGCATCCATAAGACCAACCACCTCTCGGGGCGGTAGGCACGACGCATC
GAAGCGGGAAGGCTCCGGCGCTCGGCCTGAGTCACCTCAGCAGAGTGATCTG CTGACGCTCCCAACCTTGAATAACGAAACGAAAGTCGGCCCCACGCGTGTAGG
GAGCGATCGaagtgacagtatcctctgtat-3' (SEQ ID NO. 93)
encoding nucleotide of gRNA_M3, based on M2 version, but with 34 nt bases of upper
part of stem-loop 6 removed, and four nucleotides GAAA added at truncated position
for linking: 5'-CAGTGCTGATCGATCGAAACGTCGCCTGCGATAGGCGGGAGACGCTAAACGCC CGTGGAGCATCCATAAGACCAACCACCTCTCGGGGCGGTAGGCACGACGCATC
GAAGCGGGAAGGCTCCGGCGCTCGGCCTGAGTCACCTCAGCAGAGTGATCTG CTGACGCTCCCAACCTTGAATAACGGAAACCCCACGCGTGTAGGGAGCGATCGa
agtgacagtatcctctgtat-3' (SEQ ID NO. 94)
encoding nucleotide of gRNA_M4, based on M2 version, but with 43 nt bases of upper
part of stem-loop 6 removed, and four nucleotides GAAA added at truncated position
for linking: 5'-CAGTGCTGATCGATCGAAACGTCGCCTGCGATAGGCGGGAGACGCTAAACGCC CGTGGAGCATCCATAAGACCAACCACCTCTCGGGGCGGTAGGCACGACGCATC
GAAGCGGGAAGGCTCCGGCGCTCGGCCTGAGTCACCTCAGCAGAGTGATCTG CTGACGCTCCCAACCTTGAATAACGGAAACCCCACGCGTGTAGGGAGCGATCGa
agtgacagtatcctctgtat-3' (SEQ ID NO. 95)
encoding nucleotide of gRNA_M5, based on M2 version, but with 51 nt bases of upper
part of stem-loop 6removed, and four nucleotides GAAA added at truncated position
for linking: 5'-CAGTGCTGATCGATCGAAACGTCGCCTGCGATAGGCGGGAGACGCTAAACGCC CGTGGAGCATCCATAAGACCAACCACCTCTCGGGGCGGTAGGCACGACGCATC
GAAGCGGGAAGGCTCCGGCGCTCGGCCTGAGTCACCTCAGCAGAGTGATCTG CTGACGCTCCCAACCTTGAATGAAAACGCGTGTAGGGAGCGATCGaagtgacagtatc ctctgtat-3' (SEQ ID NO. 96)
encoding nucleotide of gRNA_M6, based on M2 version, but with 57 nt bases of upper
part of stem-loop 6 removed, and four nucleotides GAAA added at truncated position
for linking: 5'-CAGTGCTGATCGATCGAAACGTCGCCTGCGATAGGCGGGAGACGCTAAACGCC CGTGGAGCATCCATAAGACCAACCACCTCTCGGGGCGGTAGGCACGACGCATC
GAAGCGGGAAGGCTCCGGCGCTCGGCCTGAGTCACCTCAGCAGAGTGATCTG CTGACGCTCCCAACCTTGGAAACGTGTAGGGAGCGATCGaagtgacagtatcctctgtat-3' (SEQ ID NO. 97)
encoding nucleotide of gRNA_M7, based on M2 version, but with 64 nt bases of upper
part of stem-loop 6 removed, and four nucleotides GAAA added at truncated position
for linking: 5'-CAGTGCTGATCGATCGAAACGTCGCCTGCGATAGGCGGGAGACGCTAAACGCC CGTGGAGCATCCATAAGACCAACCACCTCTCGGGGCGGTAGGCACGACGCATC
GAAGCGGGAAGGCTCCGGCGCTCGGCCTGAGTCACCTCAGCAGAGTGATCTG CTGACGCTCCCAACGAAAGTAGGGAGCGATCGaagtgacagtatcctctgtat- 3' (SEQ ID NO. 98)
encoding nucleotide of gRNA_M8, based on M2 version, but with 70 nt bases of upper
part of stem-loop 6 removed, and four nucleotides GAAA added at truncated position
for linking: 5'-CAGTGCTGATCGATCGAAACGTCGCCTGCGATAGGCGGGAGACGCTAAACGCC CGTGGAGCATCCATAAGACCAACCACCTCTCGGGGCGGTAGGCACGACGCATC
GAAGCGGGAAGGCTCCGGCGCTCGGCCTGAGTCACCTCAGCAGAGTGATCTG CTGACGCTCCCGAAAGGGAGCGATCGaagtgacagtatcctctgtat-3' (SEQ ID NO. 99)
encoding nucleotide of gRNA_M9, based on M2 version, but with 74 nt bases of upper
part of stem-loop 6 removed, and four nucleotides GAAA added at truncated position
for linking: 5'-CAGTGCTGATCGATCGAAACGTCGCCTGCGATAGGCGGGAGACGCTAAACGCC CGTGGAGCATCCATAAGACCAACCACCTCTCGGGGCGGTAGGCACGACGCATC
GAAGCGGGAAGGCTCCGGCGCTCGGCCTGAGTCACCTCAGCAGAGTGATCTG CTGACGCTCGAAAGAGCGATCGaagtgacagtatcctctgtat-3' (SEQ ID NO. 100)
encoding nucleotide of gRNA_M10, based on M2 version, but with 78 nt bases of upper
part of stem-loop 6 removed, and four nucleotides GAAA added at truncated position
for linking: 5'-CAGTGCTGATCGATCGAAACGTCGCCTGCGATAGGCGGGAGACGCTAAACGCC CGTGGAGCATCCATAAGACCAACCACCTCTCGGGGCGGTAGGCACGACGCATC
GAAGCGGGAAGGCTCCGGCGCTCGGCCTGAGTCACCTCAGCAGAGTGATCTG CTGACGCGAAAGCGATCGaagtgacagtatcctctgtat-3' (SEQ ID NO. 101)
encoding nucleotide of gRNA_M11, based on M2 version, but with 82 nt bases of upper
part of stem-loop 6 removed, and four nucleotides GAAA added at truncated position
for linking: 5'-CAGTGCTGATCGATCGAAACGTCGCCTGCGATAGGCGGGAGACGCTAAACGCC CGTGGAGCATCCATAAGACCAACCACCTCTCGGGGCGGTAGGCACGACGCATC
GAAGCGGGAAGGCTCCGGCGCTCGGCCTGAGTCACCTCAGCAGAGTGATCTG CTGACGCTCCCAACGAAAGTAGGGAGCGATCGaagtgacagtatcctctgtat- 3' (SEQ ID NO. 102)
encoding nucleotide of gRNA_M12, based on M1 version, but with TTTTATTTTTT added at
3' end: 5'-GAACGCGGCCCGGAACATCGAACGCCACGCAGTGCTGATCGATCGAAACGTCG CCTGCGATAGGCGGGAGACGCTAAACGCCCGTGGAGCATCCATAAGACCAACC
ACCTCTCGGGGCGGTAGGCACGACGCATCGAAGCGGGAAGGCTCCGGCGCTC GGCCTGAGTCACCTCAGCAGAGTGATCTGCTGACGCTCCCAACCTTGAATAACG
AAACGGCAACGCCTCCATAGCGGTGCAGGTCAATAAGGGTCGGCCCCACGCGT GTAGGGAGCGATCGaagtgacagtatcctctgtatTTTTATTTTTT-3' (SEQ ID NO. 103)
encoding nucleotide of gRNA_M13, based on M1 version, but with TTGACGCGGTTCTATCTAGTTACGCGTTAAACCAACTAGAAA
added at 3' end: 5'-GAACGCGGCCCGGAACATCGAACGCCACGCAGTGCTGATCGATCGAAACGTCG CCTGCGATAGGCGGGAGACGCTAAACGCCCGTGGAGCATCCATAAGACCAACC
ACCTCTCGGGGCGGTAGGCACGACGCATCGAAGCGGGAAGGCTCCGGCGCTC GGCCTGAGTCACCTCAGCAGAGTGATCTGCTGACGCTCCCAACCTTGAATAACG
AAACGGCAACGCCTCCATAGCGGTGCAGGTCAATAAGGGTCGGCCCCACGCGT GTAGGGAGCGATCGaagtgacagtatcctctgtatTTGACGCGGTTCTATCTAGTTACGCGT TAAACCAACTAGAAA-3' (SEQ ID NO. 104)
encoding nucleotide of gRNA_M14, based on M1 version, but with ACATGCGATTGACGCGGTTCTATCTAGTTACGCGTTAAACCAACTAGAAA
added at 3' end: 5'-GAACGCGGCCCGGAACATCGAACGCCACGCAGTGCTGATCGATCGAAACGTCG CCTGCGATAGGCGGGAGACGCTAAACGCCCGTGGAGCATCCATAAGACCAACC
ACCTCTCGGGGCGGTAGGCACGACGCATCGAAGCGGGAAGGCTCCGGCGCTC GGCCTGAGTCACCTCAGCAGAGTGATCTGCTGACGCTCCCAACCTTGAATAACG
AAACGGCAACGCCTCCATAGCGGTGCAGGTCAATAAGGGTCGGCCCCACGCGT GTAGGGAGCGATCGaagtgacagtatcctctgtatACATGCGATTGACGCGGTTCTATCTAG TTACGCGTTAAACCAACTAGAAA-3' (SEQ ID NO. 105)
[0144] Accordingly, the nucleotide sequences of the guide RNAs are set forth in SEQ ID NO.
4-SEQ ID NO. 17. The underlined part represents the gene targeting section, which
is a fragment with a length of 20 bp downstream of the preferred PAM sequences. In
this example, human embryonic kidney cell HEK293T is used as the experimental cell.
1. Construction of pAcC2C9hs-M1-G1 plasmid
1.1 Construction of pAcC2C9hs-M1-NSP plasmid (without gene targeting section)
[0145] The following sequences were synthesized by Sangon Biotech (Shanghai) Co., Ltd.:
expression cassette of human codon-optimized gene encoding AcC2C9: 5'-ggtgaagggggcggccgctcgaggctagtctcgtgatcgataccgtcgagatccgttcactaatcggtacctatcgat
agagaaatgttctggcacctgcacttgcactggggacagcctattttgctagtttgttttgtttcgttttgttttgatggagagc
gtatgttagtactatcgattcttgaccgacaattgcatgaagaatctgcttagggttaggcgttttgcgctgcttcgcgatgt
acgggccagatatacgcgttgacattgattattgactagttattaatagtaatcaattacggggtcattagttcatagccca
tatatggagttccgcgttacataacttacggtaaatggcccgcctggctgaccgcccaacgacccccgcccattgacgt caataatgacgtatgttcccatagtaacgccaatagggactttccattgacgtcaatgggtggagtatttacggtaaact
gcccacttggcagtacatcaagtgtatcatatgccaagtacgccccctattgacgtcaatgacggtaaatggcccgcct ggcattatgcccagtacatgaccttatgggactttcctacttggcagtacatctacgtattagtcatcgctattaccatggtg
atgcggttttggcagtacatcaatgggcgtggatagcggtttgactcacggggatttccaagtctccaccccattgacgt caatgggagtttgttttggcaccaaaatcaacgggactttccaaaatgtcgtaacaactccgccccattgacgcaaatg
ggcggtaggcgtgtacggtgggaggtctatataagcagagctggtttagtgaaccgtcagatccgctagagatccgc ggccgctaatacgactcactatagggagagccgccaccatgGCCCCTAAGAAGAAGAGGAAGGTC
ATGGTGCAGACAGAGATCCTGAAGGCCTTTCGGTTCGCCCTCGATCCTACTTCC GTCCAGGTGGCCGCCCTGAGCCGGCACGCCGGGGCCGCCAGGTGGGCCTTCA
ACCACGCCCTCGCCGCCAAGGTGGGGGCCCACGAGCGGTGGCGGGCCGAGG TGGCCAAGCTCGTGGGCGATGGCGTGCCCGAGGAGCAGGCCCGGCGGCAGG
TCCGGGTCCCCGTGCCTATGAAGCCAGCCATCCAGAAGGCCCTGAACGCCGTG AAGGGGGATTCCCGGAAGGGCCTGGATGGGGCCTGCCCATGGTGGCACGAGG
TGAACACCTACGCCTTCCAGAGCGCCTTTATTGACGCCGACCAGGCATGGAAG AACTGGCTCGACAGCCTGAGCGGCAAGAGAGTGGGGAGGAGGGTGGGCTACC
CCCGGTTTAAGAAGAAGGGCCGGGCTCGGGATAGCTTTAGATTGCACCACGAT GTGAAGAAGCCCAGCATCAGGCTGGCTGGCTACAGAAGGCTGAGGCTGCCTAG
GATTGGCGAGGTGAGGCTGCACGACAGCGGCAAGCGGCTCGCCAGGCTCATT GACAGAGGCGACGCCGTGGTGCAGTCCGTGACTGTGAGTAGGGGCGGACACA
GGTGGTACGCCTCCGTGCTCTGCAAGGTGACAGTGCAGGTGCCCGATCGGCC CAGCAGGCGGCAGCGGGAGAGGGGCGCCGTGGGAGTGGACCTGGGGGTGAA
GGTGCTGGCCGCCCTGTCCAAGCCCCTGGTCGTCGACGACCCAAGCAGCGCC CTGGTGAGAAACCCCCAGCACCTGAGGCAGGCTGAGAGGCGGCTGCTCAAGG
CCCAGCGGGCCCTGGCCAGGACACAGAAGGGCAGCGCCAGGAGAGAGAAGG CCAAGCGGCGGGTGGGGAGGGCCCATCACGAGGTGGCCGTCCGGCGGCACG
CCGCCCTGCACCAGATCACCAAGCGGCTGACCACAGGGTTCGCCGTGGTGGC CCTGGAGGATCTGAACGTCGCCGGCATGACCAGGTCCGCCAGGGGGACAGTG
GCCGCCCCAGGGAAGAACGTCCGGCAGAAGGCCGGGCTGAACCGGGTCATCC TGGACAGCGCCCCTGCCGAGCTGCGGAGGCAGGTGAACTACAAGGCCACCTG
GTACGGGAGCGAGCTGGCCGTGGCCGACAGGTGGTTCCCCTCCAGCAAGACC TGCAGCGGGTGCGGCTGGCAGAACCCCCACTTGAAGCTCTCCGATAGGGTGTT
TAGGTGCACCGACTGCGGCCTGGTCATGGATAGAGATATGAACGCCGCTAGGA ACATCGAGAGGCACGCTGTGTTGATTGATAGGAACGTGGCCTGCGACCGGCGG
GAGACTTTGAACGCCAGGGGGGCCAGCATCCGGCCCACCACAAGCCGGGGGG GGCGGCACGACGCCAGCAAGAGGGAGGGGAGCGGGGCCCGGCCAGAGTCCC
CCCAGCAGTCTGATCTGCTCACACTCCCCACACTGAACAACGAAACCGCCACAC CCCCCtctggtggttctcccaagaagaagaggaaagtctaaccggtcatcatcaccatcaccat-3'
(SEQ ID NO. 18);
expression cassette of corresponding guide RNA gRNA_M1 for AcC2C9 in human cells:
5'-gagggcctatttcccatgattccttcatatttgcatatacgatacaaggctgttagagagataattggaattaatttgactgt
aaacacaaagatattagtacaaaatacgtgacgtagaaagtaataatttcttgggtagtttgcagttttaaaattatgtttta
aaatggactatcatatgcttaccgtaacttgaaagtatttcgatttcttggctttatatatcttgtggaaaggacgaaacacc
GGAACGCGGCCCGGAACATCGAACGCCACGCAGTGCTGATCGATCGAAACGTC GCCTGCGATAGGCGGGAGACGCTAAACGCCCGTGGAGCATCCATAAGACCAAC
CACCTCTCGGGGCGGTAGGCACGACGCATCGAAGCGGGAAGGCTCCGGCGCT CGGCCTGAGTCACCTCAGCAGAGTGATCTGCTGACGCTCCCAACCTTGAATAAC
GAAACGGCAACGCCTCCATAGCGGTGCAGGTCAATAAGGGTCGGCCCCACGC GTGTAGGGAGCGATCGcgagaccattggtctcatttttttgaattctcgacctcgagacaaatggcagtattca
tccacaattttaaaagaaaaggggggattggggggtacagtgcaggggaaagaatagtagacataatagcaacag acatacaaactaaagaattacaaaaacaaattacaaaaattcaaaattttcgggtttattacagggacagcagagatc
cactttggccgcggctcgag-3' (SEQ ID NO. 19);
expression cassette of puromycin resistance gene: 5'-ggggttggggttgcgccttttccaaggcagccctgggtttgcgcagggacgcggctgctctgggcgtggttccgggaa
acgcagcggcgccgaccctgggactcgcacattcttcacgtccgttcgcagcgtcacccggatcttcgccgctaccctt gtgggccccccggcgacgcttcctgctccgcccctaagtcgggaaggttccttgcggttcgcggcgtgccggacgtga
caaacggaagccgcacgtctcactagtaccctcgcagacggacagcgccagggagcaatggcagcgcgccgac cgcgatgggctgtggccaatagcggctgctcagcagggcgcgccgagagcagcggccgggaaggggcggtgcg
ggaggcggggtgtggggcggtagtgtgggccctgttcctgcccgcgcggtgttccgcattctgcaagcctccggagcg cacgtcggcagtcggctccctcgttgaccgaatcaccgacctctctccccagggggatccaccggagcttaccatgac
cgagtacaagcccacggtgcgcctcgccacccgcgacgacgtccccagggccgtacgcaccctcgccgccgcgtt cgccgactaccccgccacgcgccacaccgtcgatccggaccgccacatcgagcgggtcaccgagctgcaagaac
tcttcctcacgcgcgtcgggctcgacatcggcaaggtgtgggtcgcggacgacggcgccgcggtggcggtctggac cacgccggagagcgtcgaagcgggggcggtgttcgccgagatcggcccgcgcatggccgagttgagcggttcccg
gctggccgcgcagcaacagatggaaggcctcctggcgccgcaccggcccaaggagcccgcgtggttcctggcca ccgtcggcgtctcgcccgaccaccagggcaagggtctgggcagcgccgtcgtgctccccggagtggaggcggccg
agcgcgccggggtgcccgccttcctggaaacctccgcgccccgcaacctccccttctacgagcggctcggcttcacc gtcaccgccgacgtcgaggtgcccgaaggaccgcgcacctggtgcatgacccgcaagcccggtgcctgacgccc
gccccacgacccgcagcgcccgaccgaaaggagcgcacgaccccatgcatcggtacctttaagaccaatgactta caaggcagctgtagatcttagccactttctagagtcggggcggccggccgcttcgagcagacatga-3'
(SEQ ID NO. 20);
human transient expression plasmid backbone: 5'-taagatacattgatgagtttggacaaaccacaactagaatgcagtgaaaaaaatgctttatttgtgaaatttgtgatgcta
ttgctttatttgtaaccattataagctgcaataaacaagttaacaacaacaattgcattcattttatgtttcaggttcaggggg
aggtgtgggaggttttttaaagcaagtaaaacctctacaaatgtggtaaaatcgataaggatccgtcgaccgatgccct tgagagccttcaacccagtcagctccttccggtgggcgcggggcatgactatcgtcgccgcacttatgactgtcttcttta
tcatgcaactcgtaggacaggtgccggcagcgctcttccgcttcctcgctcactgactcgctgcgctcggtcgttcggct gcggcgagcggtatcagctcactcaaaggcggtaatacggttatccacagaatcaggggataacgcaggaaagaa
catgtgagcaaaaggccagcaaaaggccaggaaccgtaaaaaggccgcgttgctggcgtttttccataggctccgc ccccctgacgagcatcacaaaaatcgacgctcaagtcagaggtggcgaaacccgacaggactataaagatacca
ggcgtttccccctggaagctccctcgtgcgctctcctgttccgaccctgccgcttaccggatacctgtccgcctttctccctt
cgggaagcgtggcgctttctcaatgctcacgctgtaggtatctcagttcggtgtaggtcgttcgctccaagctgggctgtg
tgcacgaaccccccgttcagcccgaccgctgcgccttatccggtaactatcgtcttgagtccaacccggtaagacacg acttatcgccactggcagcagccactggtaacaggattagcagagcgaggtatgtaggcggtgctacagagttcttga
agtggtggcctaactacggctacactagaaggacagtatttggtatctgcgctctgctgaagccagttaccttcggaaa aagagttggtagctcttgatccggcaaacaaaccaccgctggtagcggtggtttttttgtttgcaagcagcagattacgc
gcagaaaaaaaggatctcaagaagatcctttgatcttttctacggggtctgacgctcagtggaacgaaaactcacgtta agggattttggtcatgagattatcaaaaaggatcttcacctagatccttttaaattaaaaatgaagttttaaatcaatctaa
agtatatatgagtaaacttggtctgacagttaccaatgcttaatcagtgaggcacctatctcagcgatctgtctatttcgttc
atccatagttgcctgactccccgtcgtgtagataactacgatacgggagggcttaccatctggccccagtgctgcaatg ataccgcgggacccacgctcaccggctccagatttatcagcaataaaccagccagccggaagggccgagcgcag
aagtggtcctgcaactttatccgcctccatccagtctattaattgttgccgggaagctagagtaagtagttcgccagttaat
agtttgcgcaacgttgttgccattgctacaggcatcgtggtgtcacgctcgtcgtttggtatggcttcattcagctccggttcc
caacgatcaaggcgagttacatgatcccccatgttgtgcaaaaaagcggttagctccttcggtcctccgatcgttgtcag aagtaagttggccgcagtgttatcactcatggttatggcagcactgcataattctcttactgtcatgccatccgtaagatgc
ttttctgtgactggtgagtactcaaccaagtcattctgagaatagtgtatgcggcgaccgagttgctcttgcccggcgtca
atacgggataataccgcgccacatagcagaactttaaaagtgctcatcattggaaaacgttcttcggggcgaaaactc tcaaggatcttaccgctgttgagatccagttcgatgtaacccactcgtgcacccaactgatcttcagcatcttttactttcac
cagcgtttctgggtgagcaaaaacaggaaggcaaaatgccgcaaaaaagggaataagggcgacacggaaatgtt gaatactcatactcttcctttttcaatattattgaagcatttatcagggttattgtctcatgagcggatacatatttgaatgtattt
agaaaaataaacaaataggggttccgcgcacatttccccgaaaagtgccacctgcaggaaacagctatgaccatg attacgccaagctctagctagaggtcgacggtatcgagccccagctggttctttccgcctcagaagccatagagccca
ccgcatccccagcatgcctgctattgtcttcccaatcctcccccttgctgtcctgccccaccccaccccccagaatagaa tgacacctactcagacaatgcgatgcaatttcctcattttattaggaaaggacagtgggagtggcaccttccagggtca
aggaaggcacgggggaggggcaaacaacagatggctggcaactagaaggcacagtcgaggctgatcagcggg tttaaactca-3'
(SEQ ID NO. 21);
[0146] Gibson assembly technology was used to assemble these four fragments into a pAcC2C9hs-M1-NSP
plasmid. The pAcC2C9hs-M1-NSP plasmid was transformed into competent
E.
coli DH5α cells, and monoclones were picked for sequencing, and the pAcC2C9hs-M1-NSP plasmid
was obtained.
1.2 Construction of pAcC2C9hs-M1-G1 plasmid
[0147] VEGFA in HEK293T cell genome was selected as the target sequence, and a target sequence
DNA of 20 bp in length was selected. The following sequences were synthesized by Sangon
Biotech (Shanghai) Co., Ltd.:
Guide1_F: 5'-ATCGAAGTGACAGTATCCTCTGTAT-3' (SEQ ID NO. 22)
Guide1_R: 5'-AAAAATACAGAGGATACTGTCACT-3' (SEQ ID NO. 23)
Phosphorylation and annealing of target sequence DNA: the annealing systems are shown
in Table 1 below.
Table 1
| Component |
Volume |
| 10×T4 DNA ligase Buffer (NEB) |
1 µl |
| Guide-F |
2 µl |
| Guide-R |
2 µl |
| T4 PNK (NEB) |
0.5 µl |
| Sterile enzyme-free water |
4.5 µl |
[0148] The phosphorylation procedure involved: reaction at 37°C for 30 minutes, subsequently,
NaCl with a final concentration of 50 mM was added to the 10 µl reaction system, followed
by slow annealing to obtain an annealed target sequence DNA.
[0149] Then, the target sequence DNA was inserted into the pAcC2C9hs-M1-NSP plasmid by Golden
Gate assembly to construct a series of pAcC2C9hs-M1-G1 plasmids. The Golden gate assembly
system is shown in Table 2 below.
Table 2
| Component |
Content |
| 10×T4 DNA ligase Buffer (NEB) |
1 µl |
| pAcC2C9hs-M1-NSP |
50 ng |
| Targeted sequence DNA |
0.5 µl |
| T4 DNA ligase (NEB) |
0.5 µl |
| BsaI (NEB) |
0.5 µl |
| Sterile enzyme-free water |
To 10 µl |
[0150] Ligation procedure: 37°C for 2 min, followed by 16°C for 3 min, repeated for 25 cycles,
and finally incubated 80°C for 10 min.
[0151] The ligated product was transformed into E. coli DH5α cells, and monoclones were
picked for sequencing, the pAcC2C9hs-M1-G1 plasmid (a transient expression plasmid)
containing the target sequence was obtained.
2. Construction of a series of pAcC2C9hs-M2-M14-G1 plasmids
[0152] The plasmid construction was the same as Point 1 (see Construction of pAcC2C9hs-M1-G1
plasmid), except that the expression cassette of the gRNA gRNA_M1 for AcC2C9 in human
cells was replaced with the expression cassette of gRNA_M2-gRNA_M14. The 13 optimized
versions of the plasmid were named as: pAcC2C9hs-M2-G1, pAcC2C9hs-M3-G1, pAcC2C9hs-M4-G1,
pAcC2C9hs-M5-G1, pAcC2C9hs-M6-G1, pAcC2C9hs-M7-G1, pAcC2C9hs-M8-G1, pAcC2C9hs-M9-G1,
pAcC2C9hs-M10-G1, pAcC2C9hs-M11-G1, pAcC2C9hs-M12-G1, pAcC2C9hs-M13-G1, and pAcC2C9hs-M14-G1,
respectively.
3. Gene editing of human cells mediated by pAcC2C9hs-M2-M14-G1 series of plasmids
[0153] The activated HEK293T cells were cultured in DMEM medium containing 10% (by volume)
FBS. When the cell density reached about 90%, the cells were passaged into a 24-well
plate, with approximately 1.0×105 cells per well. After 16-18 hours, 1000 ng of the
pAcC2C9hs-M1-M14-G1 plasmids editing different genes was used to transfect the cells
using 2 µL of Lipofectamine 3000 (Invitrogen), respectively, in each well. After 24
hours, fresh medium containing puromycin with a final concentration of 2 µg/ml was
added for screening. After performing culture for 48 hours, the adherent cells were
digested, and genomic DNA was extracted.
[0154] The target gene fragment of the target sequence was amplified by PCR, and after gel
extraction, the PCR product was annealed using NEBuffer2 (NEB). Subsequently, T7 endonuclease
1 (NEB) was added to the PCR reaction system. followed by enzymatic digestion at 37°C
for 15 min, into which 6× Gel Loading Dye (NEB) was then added to terminate the reaction.
The reaction products were separated by 6% TBE-PAGE and stained for imaging using
4S Red dye (Sangon Biotech (Shanghai) Co., Ltd.).
[0155] 4. The gene editing results of the engineered gRNA_M1 to gRNA_M14 for AcC2C9 in mammalian
cells are shown in Fig. 2. As shown in the figure, the engineered RNAs for AcC2C9,
namely gRNA_M2, gRNA_M3, gRNA_M4, gRNA_M5, gRNA_M6, gRNA_M7, gRNA_M8, gRNA_M9, and
gRNA_M10, can improve the gene editing efficiency of AcC2C9 in mammalian cells. gRNA_M9
exhibits the highest efficiency; whereas it can be seen that adding a stabilizing
structure at the 3' end cannot improve gene editing efficiency.
Example 2 Gene editing in mammalian cells using engineered AcC2C9 nuclease gRNA
[0156] This example is based on Example 1. In this example, the engineered guide RNA gRNA_M9
(set forth in SEQ ID NO. 12) was selected for AcC2C9. In this example, human embryonic
kidney cell HEK293T was used as the experimental cell.
1. Construction of a series of pAcC2C9hs-M 1-G plasm ids
[0157] In this example, six genes, namely VEGFA, AAVS1, PDCD1, HEXA, EMX1, and TP53 in HEK293T
cell genome were selected as the target sequences, and 35 target sequence DNAs of
20 bp in length were selected from these target sequences. The 35 targetsequences
comprise G1 described in Example 1. The following sequences were synthesized by Sangon
Biotech (Shanghai) Co., Ltd.:
Guide2_F: 5'-ATCGCATGGAAACAGACCTGGCAG-3' (SEQ ID NO. 24)
Guide2_R: 5'-AAAACTGCCAGGTCTGTTTCCATG-3' (SEQ ID NO. 25)
Guide3_F: 5'-ATCGGGATGTGGTGCATTTGGAAT-3' (SEQ ID NO. 26)
Guide3_R: 5'-AAAAATTCCAAATGCACCACATCC-3' (SEQ ID NO. 27)
Guide4_F: 5'-ATCGCGACTCAACCTGGTAAACAT-3' (SEQ ID NO. 28)
Guide4_R: 5'-AAAAATGTTTACCAGGTTGAGTCG-3' (SEQ ID NO. 29)
Guide5_F: 5'-ATCGAATCATTTCCCCAAGAGGAA-3' (SEQ ID NO. 30)
Guide5_R: 5'-AAAATTCCTCTTGGGGAAATGATT-3' (SEQ ID NO. 31)
Guide6_F: 5'-ATCGAGGCAAGCATGGAAACAGAC-3' (SEQ ID NO. 32)
Guide6_R: 5'-AAAAGTCTGTTTCCATGCTTGCCT-3' (SEQ ID NO. 33)
Guide7_F: 5'-ATCGTGTTTCTGCCAGGTCTGTTT-3' (SEQ ID NO. 34)
Guide7_R: 5'-AAAAAAACAGACCTGGCAGAAACA-3' (SEQ ID NO. 35)
Guide8_F: 5'-ATCGAAGGGATGTGGTGCATTTGG-3' (SEQ ID NO. 36)
Guide8_R: 5'-AAAACCAAATGCACCACATCCCTT-3' (SEQ ID NO. 37)
Guide9_F: 5'-ATCGTGTAAGGAAGCTGCAGCACC-3' (SEQ ID NO. 38)
Guide9_R: 5'-AAAAGGTGCTGCAGCTTCCTTACA-3' (SEQ ID NO. 39)
Guide10_F: 5'-ATCGGGAGACATCCGTCGGAGAAG-3' (SEQ ID NO. 40)
Guide10_R: 5'-AAAACTTCTCCGACGGATGTCTCC-3' (SEQ ID NO. 41)
Guide11_F: 5'-ATCGAAGGATGGAGAAAGAGAAAG-3' (SEQ ID NO. 42)
Guide11_R: 5'-AAAACTTTCTCTTTCTCCATCCTT-3' (SEQ ID NO. 43)
Guide12_F: 5'-ATCGAGGAGAAGCAGTTTGGAAAA-3' (SEQ ID NO. 44)
Guide12_R: 5'-AAAATTTTCCAAACTGCTTCTCCT-3' (SEQ ID NO. 45)
Guide13_F: 5'-ATCGCAAACCTTAGAGGTTCTGGC-3' (SEQ ID NO. 46)
Guide13_R: 5'-AAAAGCCAGAACCTCTAAGGTTTG-3' (SEQ ID NO. 47)
Guide14_F: 5'-ATCGGAATCTGCCTAACAGGAGGT-3' (SEQ ID NO. 48)
Guide14_R: 5'-AAAAACCTCCTGTTAGGCAGATTC-3' (SEQ ID NO. 49)
Guide15_F: 5'-ATCGGAGAGAGATGGCTCCAGGAA-3' (SEQ ID NO. 50)
Guide15_R: 5'-AAAATTCCTGGAGCCATCTCTCTC-3' (SEQ ID NO. 51)
Guide16_F: 5'-ATCGAGGATGGAGAGGTGGCTAAA-3' (SEQ ID NO. 52)
Guide16_R: 5'-AAAATTTAGCCACCTCTCCATCCT-3' (SEQ ID NO. 53)
Guide17_F: 5'-ATCGAGCTAGCACAGACTAGAGAG-3' (SEQ ID NO. 54)
Guide17_R: 5'-AAAACTCTCTAGTCTGTGCTAGCT-3' (SEQ ID NO. 55)
Guide18_F: 5'-ATCGGCCATCCTAAGAAACGAGAG-3' (SEQ ID NO. 56)
Guide18_R: 5'-AAAACTCTCGTTTCTTAGGATGGC-3' (SEQ ID NO. 57)
Guide19_F: 5'-ATCGTACCCCGTCTCCCTGGCTTT-3' (SEQ ID NO. 58)
Guide19_R: 5'-AAAAAAAGCCAGGGAGACGGGGTA-3' (SEQ ID NO. 59)
Guide20_F: 5'-ATCGGGGGCAAAGACTGGACCCTG-3' (SEQ ID NO. 60)
Guide20_R: 5'-AAAACAGGGTCCAGTCTTTGCCCC-3' (SEQ ID NO. 61)
Guide21_F: 5'-ATCGCTCGGAGCTGGGACCACGTG-3' (SEQ ID NO. 62)
Guide21_R: 5'-AAAACACGTGGTCCCAGCTCCGAG-3' (SEQ ID NO. 63)
Guide22_F: 5'-ATCGACAGTGGGGACTAGAGCTCA-3' (SEQ ID NO. 64)
Guide22_R: 5'-AAAATGAGCTCTAGTCCCCACTGT-3' (SEQ ID NO. 65)
Guide23_F: 5'-ATCGCATCTTTGCTGTGAGCTCTA-3' (SEQ ID NO. 66)
Guide23_R: 5'-AAAATAGAGCTCACAGCAAAGATG-3' (SEQ ID NO. 67)
Guide24_F: 5'-ATCGATGCTTCAGAGACGAGATGG-3' (SEQ ID NO. 68)
Guide24_R: 5'-AAAACCATCTCGTCTCTGAAGCAT-3' (SEQ ID NO. 69)
Guide25_F: 5'-ATCGGAGCTCTACACCACACCCAA-3' (SEQ ID NO. 70)
Guide25_R: 5'-AAAATTGGGTGTGGTGTAGAGCTC-3' (SEQ ID NO. 71)
Guide26_F: 5'-ATCGCTTAAGCAAATGCCACAGCT-3' (SEQ ID NO. 72)
Guide26_R: 5'-AAAAAGCTGTGGCATTTGCTTAAG-3' (SEQ ID NO. 73)
Guide27_F: 5'-ATCGTTTAACTACTTACTGTTTGT-3' (SEQ ID NO. 74)
Guide27_R: 5'-AAAAACAAACAGTAAGTAGTTAAA-3' (SEQ ID NO. 75)
Guide28_F: 5'-ATCGCTGTGGCATTTGCTTAAGCT-3' (SEQ ID NO. 76)
Guide28_R: 5'-AAAAAGCTTAAGCAAATGCCACAG-3' (SEQ ID NO. 77)
Guide29_F: 5'-ATCGGTTGAACAAACAGTAAGTAG-3' (SEQ ID NO. 78)
Guide29_R: 5'-AAAACTACTTACTGTTTGTTCAAC-3' (SEQ ID NO. 79)
Guide30_F: 5'-ATCGAGAGATGCCTTACTAGGTAC-3' (SEQ ID NO. 80)
Guide30_R: 5'-AAAAGTACCTAGTAAGGCATCTCT-3' (SEQ ID NO. 81)
Guide31_F: 5'-ATCGAGCTTTACAAAAGAAGGGGA-3' (SEQ ID NO. 82)
Guide31_R: 5'-AAAATCCCCTTCTTTTGTAAAGCT-3' (SEQ ID NO. 83)
Guide32_F: 5'-ATCGGACAAGGTTGAACAAACAGT-3' (SEQ ID NO. 84)
Guide32_R: 5'-AAAAACTGTTTGTTCAACCTTGTC-3' (SEQ ID NO. 85)
Guide33_F: 5'-ATCGCAGCTCATTTACAGACGGGA-3' (SEQ ID NO. 86)
Guide33_R: 5'-AAAATCCCGTCTGTAAATGAGCTG-3' (SEQ ID NO. 87)
Guide34_F: 5'-ATCGGTGTCAGTTTAAGAATGGTG-3' (SEQ ID NO. 88)
Guide34_R: 5'-AAAACACCATTCTTAAACTGACAC-3' (SEQ ID NO. 89)
Guide35_F: 5'-ATCGGGTGCAGTTATGCCTCAGAT-3' (SEQ ID NO. 90)
Guide35_R: 5'-AAAAATCTGAGGCATAACTGCACC-3' (SEQ ID NO. 91)
[0158] The plasmid construction was the same as Point 1 in Example 1 (see Construction of
pAcC2C9hs-M1-G1 plasmid), using the Golden Gate assembly method to insert the annealed
double-stranded DNA into the pAcC2C9hs-M1-NSP plasmid to obtain 35 pAcC2C9hs-M1-G
plasmids.
3. Construction of a series of pAcC2C9hs-M9-G plasmids
[0159] The expression cassette of the guide RNA gRNA_M1 for AcC2C9 in human cells was replaced
with the expression cassette of the guide gRNA_M9. According to the above method,
different target sequences were inserted to obtain 35 pAcC2C9hs-M9-G series of plasmids
containing the target sequences.
4. Gene editing of human cells mediated by pAcC2C9hs-M9-G series of plasmids
[0160] The operation was the same as Point 3 in Example 1 (see Gene editing of human cells
mediated by pAcC2C9hs-M2-M14-G1 series of plasmids), except that the pAcC2C9hs-M1-M14-G1
series plasmids in Example 1 were replaced with the pAcC2C9hs-M1-G series plasmids
and the pAcC2C9hs-M9-G series plasmids.
5. Result
[0161] The editing efficiencies of gRNA_M1 and gRNA_M9 in HEK293T cells are shown in Fig.
3. As shown in the figure, using the engineered gRNA_M9 can achieve high-efficiency
editing in mammalian cells, wherein an editing efficiency at the same site of gRNA_M9
is approximately 10 times higher than that of gRNA_M1. For example, the editing efficiency
at the Guide2 site can be improved from 3% to 30%. The gene editing efficiency of
AcC2C9 in mammalian cells has been increased up to 85%, significantly improving the
activity and versatility of gene editing of AcC2C9 in mammalian cells. Additionally,
no gene editing activity is detected at the Guide6, Guide8, and Guide23 sites when
using gRNA_M1, whereas a gene editing efficiency of approximately 10% is detected
when using the engineered gRNA_M9. The engineered gRNA_M9 expands the editing scope
of the CRISPR/AcC2C9 gene editing system.
[0162] The above examples are intended to illustrate the disclosed implementation examples
of the present disclosure, and should not be construed as limiting the present disclosure.
In addition, various modifications listed herein and changes in methods in the present
disclosure are obvious to those skilled in the art without departing from the scope
and spirit of the present disclosure. Although the present disclosure has been described
in detail with reference to multiple specific preferred examples, it should be understood
that the present disclosure should not be limited to these specific examples. In fact,
various modifications as mentioned above that are obvious to those skilled in the
art to obtain the present disclosure should be included in the scope of the present
disclosure.
1. A guide RNA, comprising an RNA framework and a gene targeting section,
wherein the RNA framework comprises a tracrRNA and a tracr duplex linked directly
or through a linker;
a nucleotide sequence of the tracrRNA is a sequence obtained by addition, deletion,
or substitution of one or more nucleotides in a nucleotide sequence set forth in SEQ
ID NO. 117, and
a nucleotide sequence of the tracr duplex is a sequence obtained by addition, deletion,
or substitution of one or more nucleotides in a nucleotide sequence set forth in SEQ
ID NO. 127.
2. The guide RNA according to claim 1, wherein the linker comprises oligonucleotides;
preferably, the linker comprises 3-18 nt oligonucleotides; further preferably, a nucleotide
sequence of the linker is GAAA.
3. The guide RNA according to claim 1, wherein the gene targeting section further comprises
one or more of the following:
1) the gene targeting section is located at a 3' end of the RNA framework;
2) the gene targeting section identifies a PAM sequence on a target gene; preferably,
the PAM sequence is 5'-NAAN, wherein N is A, T, C, or G; and
3) the gene targeting section targets a nucleic acid fragment with a length of 12-40
bp downstream of the PAM sequence.
4. The guide RNA according to claim 1, wherein the guide RNA comprises stem-loop structures;
preferably, the guide RNA comprises five stem-loop structures, which are stem-loop
2, stem-loop 3, stem-loop 4, stem-loop 5 and stem-loop 6, respectively.
5. The guide RNA according to claim 1, wherein the RNA framework comprises a nucleotide
sequence obtained by addition, deletion, or substitution of 15-150 nucleotides at
a 5' or 3' end of a nucleotide sequence set forth in SEQ ID NO. 106; or a nucleotide
sequence obtained by linking sequences being truncated or substituted 15-150 nucleotides
in the middle of the nucleotide sequence set forth in SEQ ID NO. 106 through a linker.
6. The guide RNA according to claim 1, wherein the RNA framework is a gRNA_M2 RNA framework
obtained by deletion of 30 bases in stem-loop 1 in the RNA framework with a nucleotide
sequence set forth in SEQ ID NO. 106; preferably, a nucleotide sequence of a tracrRNA
in the gRNA_M2 RNA framework is set forth in SEQ ID NO. 140, and/or, a nucleotide
sequence of a tracr duplex in the gRNA_M2 RNA framework is set forth in SEQ ID NO.
127; more preferably, a nucleotide sequence of the gRNA_M2 RNA framework is set forth
in SEQ ID NO. 107.
7. The guide RNA according to claim 1, wherein the RNA framework is a gRNA_M3 RNA framework
obtained by deletion of 34 bases in stem-loop 6 in the RNA framework with a nucleotide
sequence set forth in SEQ ID NO. 107; preferably, a nucleotide sequence of a tracrRNA
in the gRNA_M3 RNA framework is set forth in SEQ ID NO. 118, and/or, a nucleotide
sequence of a tracr duplex in the gRNA_M3 RNA framework is set forth in SEQ ID NO.
128; more preferably, a nucleotide sequence of the gRNA_M3 RNA framework is set forth
in SEQ ID NO. 108.
8. The guide RNA according to claim 1, wherein the RNA framework is a gRNA_M4 RNA framework
obtained by deletion of 43 bases in stem-loop 6 in the RNA framework with a nucleotide
sequence set forth in SEQ ID NO. 107; preferably, a nucleotide sequence of a tracrRNA
in the gRNA_M4 RNA framework is set forth in SEQ ID NO. 119, and/or, a nucleotide
sequence of a tracr duplex in the gRNA_M4 RNA framework is set forth in SEQ ID NO.
129; more preferably, a nucleotide sequence of the gRNA_M4 RNA framework is set forth
in SEQ ID NO. 109.
9. The guide RNA according to claim 1, wherein the RNA framework is a gRNA_M5 RNA framework
obtained by deletion of 51 bases in stem-loop 6 in the RNA framework with a nucleotide
sequence set forth in SEQ ID NO. 107; preferably, a nucleotide sequence of a tracrRNA
in the gRNA_M5 RNA framework is set forth in SEQ ID NO. 120, and/or, a nucleotide
sequence of a tracr duplex in the gRNA_M5 RNA framework is set forth in SEQ ID NO.
130; more preferably, a nucleotide sequence of the gRNA_M5 RNA framework is set forth
in SEQ ID NO. 110.
10. The guide RNA according to claim 1, wherein the RNA framework is a gRNA_M6 RNA framework
obtained by deletion of 57 bases in stem-loop 6 in the RNA framework with a nucleotide
sequence set forth in SEQ ID NO. 107; preferably, a nucleotide sequence of a tracrRNA
in the gRNA_M6 RNA framework is set forth in SEQ ID NO. 121, and/or, a nucleotide
sequence of a tracr duplex in the gRNA_M6 RNA framework is set forth in SEQ ID NO.
131; more preferably, a nucleotide sequence of the gRNA_M6 RNA framework is set forth
in SEQ ID NO. 111.
11. The guide RNA according to claim 1, wherein the RNA framework is a gRNA_M7 RNA framework
obtained by deletion of 64 bases in stem-loop 6 in the RNA framework with a nucleotide
sequence set forth in SEQ ID NO. 107; preferably, a nucleotide sequence of a tracrRNA
in the gRNA_M7 RNA framework is set forth in SEQ ID NO. 122, and/or, a nucleotide
sequence of a tracr duplex in the gRNA_M7 RNA framework is set forth in SEQ ID NO.
132; more preferably, a nucleotide sequence of the gRNA_M7 RNA framework is set forth
in SEQ ID NO. 112.
12. The guide RNA according to claim 1, wherein the RNA framework is a gRNA_M8 RNA framework
obtained by deletion of 70 bases in stem-loop 6 in the RNA framework with a nucleotide
sequence set forth in SEQ ID NO. 107; preferably, a nucleotide sequence of a tracrRNA
in the gRNA_M8 RNA framework is set forth in SEQ ID NO. 123, and/or, a nucleotide
sequence of a tracr duplex in the gRNA_M8 RNA framework is set forth in SEQ ID NO.
133; more preferably, a nucleotide sequence of the gRNA_M8 RNA framework is set forth
in SEQ ID NO. 113.
13. The guide RNA according to claim 1, wherein the RNA framework is a gRNA_M9 RNA framework
obtained by deletion of 74 bases in stem-loop 6 in the RNA framework with a nucleotide
sequence set forth in SEQ ID NO. 107; preferably, a nucleotide sequence of a tracrRNA
in the gRNA_M9 RNA framework is set forth in SEQ ID NO. 124, and/or, a nucleotide
sequence of a tracr duplex in the gRNA_M9 RNA framework is set forth in SEQ ID NO.
134; more preferably, a nucleotide sequence of the gRNA_M9 RNA framework is set forth
in SEQ ID NO. 114.
14. The guide RNA according to claim 1, wherein the RNA framework is a gRNA_M10 RNA framework
obtained by deletion of 78 bases in stem-loop 6 in the RNA framework with a nucleotide
sequence set forth in SEQ ID NO. 107; preferably, a nucleotide sequence of a tracrRNA
in the gRNA_M10 RNA framework is set forth in SEQ ID NO. 125, and/or, a nucleotide
sequence of a tracr duplex in the gRNA_M10 RNA framework is set forth in SEQ ID NO.
135; more preferably, a nucleotide sequence of the gRNA_M10 RNA framework is set forth
in SEQ ID NO. 115.
15. The guide RNA according to claim 1, wherein the RNA framework is a gRNA_M11 RNA framework
obtained by deletion of 82 bases in stem-loop 6 in the RNA framework with a nucleotide
sequence set forth in SEQ ID NO. 107; preferably, a nucleotide sequence of a tracrRNA
in the gRNA_M11 RNA framework is set forth in SEQ ID NO. 126, and/or, a nucleotide
sequence of a tracr duplex in the gRNA_M11 RNA framework is set forth in SEQ ID NO.
136; more preferably, a nucleotide sequence of the gRNA_M11 RNA framework is set forth
in SEQ ID NO. 116.
16. The guide RNA according to claim 1, further comprising an RNA structure stabilizing
sequence provided at a 3' end of the guide RNA; preferably, the RNA structure stabilizing
sequence is provided at a 3' end of the gene targeting section.
17. The guide RNA according to claim 16, wherein the RNA structure stabilizing sequence
is set forth in any one of SEQ ID NOs. 137-139.
18. The guide RNA according to claim 1, wherein a nucleotide sequence of the guide RNA
is set forth in any one of SEQ ID NOs. 5-17.
19. An isolated polynucleotide, encoding the guide RNA according to any one of claims
1 to 18.
20. A construct, comprising the isolated polynucleotide according to claim 19.
21. An expression system, comprising the construct according to claim 20 or a genome exogenously
being introduced the polynucleotide according to claim 19.
22. A gene editing system, comprising the guide RNA according to any one of claims 1 to
18 or the polynucleotide according to claim 19.
23. The gene editing system according to claim 22, further comprising a nuclease or a
polynucleotide encoding the same.
24. The gene editing system according to claim 23, wherein the nuclease is a CRISPR nuclease;
preferably, the nuclease is selected from Cas9, Cas12 and Cas13 protein families and
variants thereof; further preferably, the Cas nuclease is selected from nSpCas9 and
a variant thereof, SaCas9 and a variant thereof, Cas12a and a variant thereof, and
C2C9 and a variant thereof; more preferably, the Cas nuclease is an AcC2C9 nuclease
or a variant thereof.
25. The gene editing system according to claim 24, wherein the AcC2C9 nuclease comprises
at least one of the following:
(I) a wild-type AcC2C9 nuclease or a fragment thereof; wherein the AcC2C9 nuclease
is derived from Actinomadura craniellae C2C9, with an amino acid sequence set forth in SEQ ID NO. 1; a human codon-optimized
nucleic acid sequence of the AcC2C9 nuclease is set forth in SEQ ID NO. 3;
or (II) except for (I), the AcC2C9 nuclease further comprises a nuclear localization
signal fragment.
26. A pharmaceutical composition, comprising the gene editing system according to any
one of claims 22 to 24, and a pharmaceutically acceptable carrier.
27. A gene editing method, comprising contacting a target gene with the gene editing system
according to any one of claims 22 to 24 to realize editing of the target gene.
28. The gene editing method according to claim 27, comprising the following:
i) introducing the AcC2C9 nuclease or the polynucleotide encoding the same and the
guide RNA or the polynucleotide encoding the same into a cell;
ii) under the mediation of the AcC2C9 nuclease, producing one or more nicks in the
target gene, or targeting, editing, modifying or manipulating the target gene.
29. The gene editing method according to claim 27 or 28, comprising the following:
1) guiding the AcC2C9 nuclease to the target gene by the guide RNA according to any
one of claims 1 to 18;
2) forming a complex comprising the AcC2C9 nuclease and the guide RNA according to
any one of claims 1 to 18, and recognizing a PAM sequence on the target gene; and
3) introducing a donor template comprising a heterologous polynucleotide sequence
into the cell.
30. Use of the guide RNA according to any one of claims 1 to 18, the isolated polynucleotide
according to claim 19, the construct according to claim 20, the expression system
according to claim 21, the gene editing system according to any one of claims 22 to
25, the pharmaceutical composition according to claim 26 or the method according to
any one of claims 27 to 29 in gene editing of a target gene and/or a related polypeptide
in an in vivo, ex vivo cell or cell-free environment.
31. The use according to claim 30, wherein the gene editing is selected from gene cleavage,
gene deletion, gene insertion, point mutation, transcription repression, transcription
activation and/or base editing.
32. A cell, obtained by gene editing with the gene editing system according to any one
of claims 21 to 24 or the method according to any one of claims 26 to 28.
33. A method for preparing a guide RNA, comprising modification of an RNA framework of
an original guide RNA, and the modification is selected from modification of tracrRNA
or crRNA in the RNA framework alone or modification of tracrRNA and crRNA in the RNA
framework simultaneously, and the modification is addition, deletion, or substitution
of one or more nucleotides in a nucleotide of a tracrRNA and/or a crRNA, or linking
the tracrRNA and the crRNA through a linker.